WO2020051169A1 - Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor - Google Patents

Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor Download PDF

Info

Publication number
WO2020051169A1
WO2020051169A1 PCT/US2019/049419 US2019049419W WO2020051169A1 WO 2020051169 A1 WO2020051169 A1 WO 2020051169A1 US 2019049419 W US2019049419 W US 2019049419W WO 2020051169 A1 WO2020051169 A1 WO 2020051169A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystalline form
compound
terms
ray powder
diffraction pattern
Prior art date
Application number
PCT/US2019/049419
Other languages
English (en)
French (fr)
Inventor
Brent Douty
David M. Burns
Andrew P. Combs
Zhongjiang JIA
Daniel Levy
Eddy W. Yue
Original Assignee
Incyte Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CR20210165A priority Critical patent/CR20210165A/es
Priority to FIEP19858443.5T priority patent/FI3847175T3/fi
Application filed by Incyte Corporation filed Critical Incyte Corporation
Priority to MX2021002551A priority patent/MX2021002551A/es
Priority to EP24153948.5A priority patent/EP4338801A3/en
Priority to CA3111629A priority patent/CA3111629A1/en
Priority to BR112021004094-3A priority patent/BR112021004094A2/pt
Priority to CN201980071241.6A priority patent/CN113677683A/zh
Priority to EP19858443.5A priority patent/EP3847175B1/en
Priority to PE2021000289A priority patent/PE20211815A1/es
Priority to EA202190679A priority patent/EA202190679A1/ru
Priority to JP2021512576A priority patent/JP2021535182A/ja
Priority to AU2019336675A priority patent/AU2019336675A1/en
Priority to RS20240361A priority patent/RS65334B1/sr
Priority to SI201930734T priority patent/SI3847175T1/sl
Priority to IL309869A priority patent/IL309869A/en
Priority to SG11202102224UA priority patent/SG11202102224UA/en
Priority to KR1020217009964A priority patent/KR20210091121A/ko
Priority to HRP20240252TT priority patent/HRP20240252T1/hr
Priority to LTEPPCT/US2019/049419T priority patent/LT3847175T/lt
Priority to DK19858443.5T priority patent/DK3847175T3/da
Publication of WO2020051169A1 publication Critical patent/WO2020051169A1/en
Priority to IL281262A priority patent/IL281262A/en
Priority to PH12021550472A priority patent/PH12021550472A1/en
Priority to CONC2021/0004061A priority patent/CO2021004061A2/es

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present invention relates to salts and crystalline forms of 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, 8- amino-N-(2-hydroxy-2-methylpropyl)-3-(2-methyl-5-( 1,1, 1 -trifluoro-2-hydroxypropan-2- yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide and 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2- (methyl-d 3 )-5 -( 1 , 1 , 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2-a]pyrazine-6-carboxamide which are PI3K inhibitors useful in the treatment of cancer and other diseases.
  • PI3K phosphoinositide 3-kinase
  • the salts and crystalline forms of the compounds of Formula A, Formula B and Formula C provided herein address the ongoing need for the development of PI3K inhibitors for the treatment of serious diseases.
  • the present invention provides salts and crystalline forms of the compound of Formula A, crystalline forms of the compound of Formula B and a crystalline form of the compound of Formula C:
  • the present invention further provides a composition comprising the salts and crystalline forms of the compound of Formula A provided herein and at least one pharmaceutically acceptable carrier.
  • the present invention further provides a composition comprising a crystalline form of the compound of Formula B and at least one pharmaceutically acceptable carrier.
  • the present invention further provides a composition comprising a crystalline form of the compound of Formula C and at least one
  • the present invention further provides a process for preparing salts and crystalline forms of the invention.
  • the present invention further provides a method of treating a disease associated with abnormal expression or activity of a PI3K kinase in a patient, comprising administering to the patient a therapeutically effective amount of a salt or crystalline form of the invention.
  • Figure 1 shows an XRPD pattern for 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3- yl)-4-methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide, crystalline Form IA.
  • Figure 2 shows the results of a DSC experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IA.
  • Figure 3 shows the results of a TGA experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IA.
  • Figure 4 shows an XRPD pattern for 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3- yl)-4-methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide, crystalline Form IIA.
  • Figure 5 shows the results of a DSC experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IIA.
  • Figure 6 shows the results of a TGA experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IIA.
  • Figure 7 shows an XRPD pattern for 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3- yl)-4-methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide, crystalline Form IIIA.
  • Figure 8 shows the results of a DSC experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IIIA.
  • Figure 9 shows the results of a TGA experiment for 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, crystalline Form IIIA
  • Figure 10 shows an XRPD pattern for 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-methyl-5- (1, 1,1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form
  • Figure 11 shows the results of a DSC experiment for 8-amino-N-(2-hydroxy-2-methylpropyl)-3- (2-methyl-5-( 1,1, 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form IB.
  • Figure 12 shows an XRPD pattern for 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-methyl-5- (1, 1,1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form IIB.
  • Figure 13 shows the results of a DSC experiment for 8-amino-N-(2-hydroxy-2-methylpropyl)-3- (2-methyl-5-( 1,1, 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form IIB.
  • Figure 14 shows an XRPD pattern for 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-(methyl-d3)- 5 -( 1 , 1 , 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form
  • Figure 15 shows the results of a DSC experiment for 8-amino-N-(2-hydroxy-2-methylpropyl)-3- (2-(methyl-d 3 )-5 -( 1,1, 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2-a]pyrazine-6-carboxamide, crystalline Form IC.
  • Figure 16A shows the asymmetric crystalline unit of 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide hydrobromic acid salt, methanol solvate form, with thermal ellipsoids drawn to the 30% probability level.
  • Figure 16B shows a crystalline unit of 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3- yl)-4-methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide hydrobromic acid salt, with thermal ellipsoids drawn to the 30% probability level.
  • the present invention relates to, inter alia, salts and crystalline forms of the PI3K inhibitor 2-(3- (8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)-3,3,3-trifluoro-2- hydroxypropanamide having Formula A:
  • the present invention also relates to, inter alia, crystalline forms of the PI3K inhibitor 8-amino- N-(2-hydroxy-2-methylpropyl)-3 -(2 -methyl-5 -(1,1,1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [1,2- a]pyrazine-6-carboxamide having Formula B:
  • the present invention also relates to, inter alia, crystalline forms of the PI3K inhibitor 8-amino- N-(2-hydroxy-2-methylpropyl)-3-(2-(methyl-d 3 )-5-( 1,1,1 -trifluoro-2-hydroxypropan-2- yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide having Formula C:
  • crystalline forms of the same substance have different bulk properties relating to, for example, hygroscopicity, solubility, stability, and the like.
  • Forms with high melting points often have good thermodynamic stability which is advantageous in prolonging shelf-life drug formulations containing the solid form.
  • Forms with lower melting points often are less thermodynamically stable, but are advantageous in that they have increased water solubility, translating to increased drug bioavailability.
  • Forms that are weakly hygroscopic are desirable for their stability to heat and humidity and are resistant to degradation during long storage.
  • Anhydrous forms are often desirable because they can be consistently made without concern for variation in weight or composition due to varying solvent or water content.
  • hydrated or solvated forms can be advantageous in that they are less likely to be hygroscopic and may show improved stability to humidity under storage conditions.
  • crystalline form is meant to refer to a certain lattice configuration of a crystalline substance.
  • Different crystalline forms of the same substance typically have different crystalline lattices (e.g., unit cells) which are attributed to different physical properties that are characteristic of each of the crystalline forms.
  • different lattice configurations have different water or solvent content.
  • the different crystalline lattices can be identified by solid state characterization methods such as by X-ray powder diffraction (XRPD). Other characterization methods such as differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), dynamic vapor sorption (DVS), solid state NMR, and the like further help identify the crystalline form as well as help determine stability and solvent/water content.
  • Crystalline forms of a substance include both solvated (e.g., hydrated) and non-solvated (e.g., anhydrous) forms.
  • a hydrated form is a crystalline form that includes water in the crystalline lattice. Hydrated forms can be stoichiometric hydrates, where the water is present in the lattice in a certain water/molecule ratio such as for hemihydrates, monohydrates, dihydrates, etc. Hydrated forms can also be non-stoichiometric, where the water content is variable and dependent on external conditions such as humidity.
  • Crystalline forms are most commonly characterized by XRPD.
  • An XRPD pattern of reflections (peaks) is typically considered a fingerprint of a particular crystalline form. It is well known that the relative intensities of the XRPD peaks can widely vary depending on, inter alia, the sample preparation technique, crystal size distribution, filters, the sample mounting procedure, and the particular instrument employed. In some instances, new peaks may be observed or existing peaks may disappear, depending on the type of instrument or the settings (for example, whether a Ni filter is used or not).
  • the term“peak” refers to a reflection having a relative height/intensity of at least about 4% of the maximum peak height/intensity.
  • instrument variation and other factors can affect the 2-theta values.
  • peak assignments such as those reported herein, can vary by plus or minus about 0.2° (2 -theta), and the term“substantially” as used in the context of XRPD herein is meant to encompass the above- mentioned variations.
  • temperature readings in connection with DSC, TGA, or other thermal experiments can vary about ⁇ 4 °C depending on the instrument, particular settings, sample preparation, etc.
  • DSC it is known that the temperatures observed will depend on the rate of the temperature change as well as the sample preparation technique and the particular instrument employed.
  • the values reported herein related to DSC thermograms can vary, as indicated above, by ⁇ 4 0 C. Accordingly, a crystalline form reported herein having a DSC thermogram“substantially” as shown in any of the Figures is understood to accommodate such variation.
  • the compound of Formula A can be isolated in numerous crystalline forms, including, for example, crystalline forms which are anhydrous, and/or non-solvated or solvated. In some embodiments, the crystalline forms of the compound of Formula A are solvated. In some embodiments, the crystalline forms of the compound of Formula A are anhydrous. In some embodiments, the crystalline forms of the compound of Formula A are non-solvated. In some embodiments, the crystalline forms of the compound of Formula A are anhydrous and non-solvated. By“anhydrous” is meant that the crystalline form of the compound of Formula A contains essentially no bound water in the crystal lattice structure, i.e.. the compound does not form a crystalline hydrate.
  • the present application provides a process of preparing a crystalline form of the compound 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)-3,3,3- trifluoro-2-hydroxypropanamide.
  • the process comprises dissolving 2-(3-(8-amino- 6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide in a solvent to form a mixture and crystallizing the compound from the mixture.
  • the solvent comprises isopropyl acetate. In some embodiments, the solvent further comprises heptane.
  • the solvent comprises methanol. In some embodiments, the process further comprises heating the mixture to a temperature of from about 70°C to about 90°C.
  • the process further comprises heating the mixture to a temperature of from about 50°C to about 70°C.
  • the process further comprises cooling the mixture to room temperature.
  • the present application further provides a crystalline form of the compound 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide, which is prepared according to a process provided herein.
  • the crystalline form is Form IA as described herein.
  • the crystalline form is Form IIA as described herein.
  • the crystalline form is Form IIIA as described herein.
  • the present application provides a process of preparing a hydrobromic acid salt of the compound 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)- 3,3,3-trifluoro-2-hydroxypropanamide.
  • the process comprises dissolving the compound of Formula A in a solvent to form a mixture and adding hydrobromic acid to the mixture.
  • the solvent comprises methanol.
  • the hydrobromic acid is added to the mixture as an aqueous solution of hydrobromic acid.
  • an excess amount of hydrobromic acid is added to the mixture based on 1 equivalent of 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide .
  • hydrobromic acid salt In some embodiments of the process of preparing the hydrobromic acid salt, about 1.1 to about 1.5 equivalents of hydrobromic acid are added to the mixture based on 1 equivalent of the 2-(3-(8-amino- 6-(trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide .
  • the process further comprises substantially isolating the 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4- methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide hydrobromic acid salt.
  • the 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide hydrobromic acid salt is isolated as a crystalline form.
  • the 2-(3-(8-amino-6- (trifluoromethyl)imidazo [ 1 ,2-a]pyrazin-3 -yl)-4-methylphenyl)-3 ,3 ,3 -trifluoro-2-hydroxypropanamide hydrobromic acid salt is isolated as a methanol solvate crystalline form.
  • the present application further provides a hydrobromic acid salt of 2-(3-(8- amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)-3,3,3-trifluoro-2- hydroxypropanamide, which is prepared according to a process provided herein.
  • the hydrobromic acid salt is crystalline.
  • the hydrobromic acid salt is a solvated crystalline form.
  • the hydrobromic acid salt is a methanol solvate crystalline form.
  • the crystalline forms of the invention are substantially isolated.
  • substantially isolated is meant that a particular crystalline form of the compound of Formula A is at least partially isolated from impurities.
  • a crystalline form of the invention comprises less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 2.5%, less than about 1%, or less than about 0.5% of impurities.
  • Impurities generally include anything that is not the substantially isolated crystalline form including, for example, other crystalline forms and other substances.
  • a crystalline form of the compound of Formula A is substantially free of other crystalline forms.
  • the phrase“substantially free of other crystalline forms” means that a particular crystalline form of the compound of Formula A comprises greater than about 80%, greater than about 90%, greater than about 95%, greater than about 98%, greater than about 99% or greater than about 99.5% by weight of the particular crystalline form.
  • the crystalline form of the compound of Formula A is Form IA. In some embodiments, crystalline Form IA of the compound of Formula A is anhydrous and non-solvated.
  • the preparation of the compound of Form IA of the compound of Formula A is described in Example 1. Crystalline Form IA of the compound of Formula A can be generally prepared as described in Example 2.
  • Crystalline Form IA of the compound of Formula A can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), and thermogravimetric analysis (TGA).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • crystalline Form IA of the compound of Formula A is characterized by an XRPD pattern substantially as shown in Figure 1. Peaks from the XRPD pattern are listed in Table 1.
  • crystalline Form IA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 8.6° ⁇ 0.2°. In some embodiments, crystalline Form IA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 9.5° ⁇ 0.2°. In some embodiments, crystalline Form IA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 10.3° ⁇ 0.2°. In some embodiments, crystalline Form IA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 14.9° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 8.6° ⁇ 0.2°; 9.5° ⁇ 0.2°; 10.3° ⁇ 0.2°; 13.0° ⁇ 0.2°; 13.6° ⁇ 0.2°; 14.2° ⁇ 0.2°; and 14.9° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 8.6° ⁇ 0.2°; 9.5° ⁇ 0.2°; 10.3° ⁇ 0.2°; 14.9° ⁇ 0.2°; 17.3° ⁇ 0.2°; 17.8° ⁇ 0.2°; 19.0° ⁇ 0.2°; 19.2° ⁇ 0.2°; 20.1° ⁇ 0.2°; 20.6° ⁇ 0.2°; 21.2° ⁇ 0.2°; 22.2 ° ⁇ 0.2°; 24.0° ⁇ 0.2°; 26.8° ⁇ 0.2°; and 28.7 ° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 10.3° ⁇ 0.2°; 14.9° ⁇ 0.2°; 17.3° ⁇ 0.2°; 19.2° ⁇ 0.2°; and 24.0° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 8.6° ⁇ 0.2°; 9.5° ⁇ 0.2°; 10.3° ⁇ 0.2°; 13.0° ⁇ 0.2°; 13.6° ⁇ 0.2°; 14.2° ⁇ 0.2°; 14.9° ⁇ 0.2°; 17.3° ⁇ 0.2°; 19.2° ⁇ 0.2°; 20.6° ⁇ 0.2°; 24.0° ⁇ 0.2°; and 28.7° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 8.6° ⁇ 0.2°; 9.5° ⁇ 0.2°; 10.3° ⁇ 0.2°; 14.9° ⁇ 0.2°; 17.3° ⁇ 0.2°; 17.8° ⁇ 0.2°; 19.0° ⁇ 0.2°; 19.2° ⁇ 0.2°; 20.1° ⁇ 0.2°; 20.6° ⁇ 0.2°; 21.2° ⁇ 0.2°; 22.2 ° ⁇ 0.2°; 24.0° ⁇ 0.2°; 26.8° ⁇ 0.2°; and 28.7 ° ⁇ 0.2°.
  • crystalline Form IA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 9.5° ⁇ 0.2°; 10.3° ⁇ 0.2°; 14.9° ⁇ 0.2°; 17.3° ⁇ 0.2°; 19.2° ⁇ 0.2°; 20.6° ⁇ 0.2°; 24.0° ⁇ 0.2°; and 28.7° ⁇ 0.2°.
  • Form IA of the compound of Formula A is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 193 °C.
  • crystalline Form IA of the compound of Formula A has a DSC thermogram substantially as shown in Figure 2.
  • crystalline Form IA of the compound of Formula A has a TGA trace substantially as shown in Figure 3.
  • the crystalline form of the compound of Formula A is Form IIA. In some embodiments, crystalline Form IIA of the compound of Formula A is anhydrous and non-solvated. This crystalline form can be generally prepared as described in Example 3.
  • Crystalline Form IIA of the compound of Formula A can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), and thermogravimetric analysis (TGA).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern substantially as shown in Figure 4. Peaks from the XRPD pattern are listed in Table 2.
  • crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 9.1 ⁇ 0.2°. In some embodiments, crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 11.1° ⁇ 0.2°. In some embodiments, crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 21.9° ⁇ 0.2°. In some
  • crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 12.6° ⁇ 0.2°. In some embodiments, crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 13.6° ⁇ 0.2°. In some embodiments, crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 18.0° ⁇ 0.2°. In some embodiments, crystalline Form IIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 20, at 19.0° ⁇ 0.2°.
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 9.1° ⁇ 0.2°; 11.1° ⁇ 0.2°; 12.6° ⁇ 0.2°; and 13.5° ⁇ 0.2°.
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 9.1° ⁇ 0.2°; 11.1° ⁇ 0.2°; 12.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 16.1° ⁇ 0.2°; 16.9° ⁇ 0.2°; 18.0° ⁇ 0.2°; 18.4° ⁇ 0.2°; 19.0° ⁇ 0.2°; 19.7° ⁇ 0.2°; 20.1° ⁇ 0.2°; 20.5° ⁇ 0.2°; 21.9° ⁇
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 20: 12.6° ⁇ 0.2°; 18.0° ⁇ 0.2°; 19.0° ⁇ 0.2°; and 21.9° ⁇ 0.2°.
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 9.1° ⁇ 0.2°; 11.1° ⁇ 0.2°; 12.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 18.0° ⁇ 0.2°; 19.0° ⁇ 0.2°; 20.5° ⁇ 0.2°; and 21.9° ⁇ 0.2°.
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 9.1° ⁇ 0.2°; 11.1° ⁇ 0.2°; 12.6° ⁇
  • crystalline Form IIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 12.6° ⁇ 0.2°; 18.0° ⁇ 0.2°; 19.0° ⁇ 0.2°; 20.5° ⁇ 0.2°; and 21.9° ⁇ 0.2°.
  • Form IIA of the compound of Formula A is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 180 °C.
  • crystalline Form IIA of the compound of Formula A has a DSC thermogram substantially as shown in Figure 5.
  • crystalline Form IIA of the compound of Formula A has a TGA trace substantially as shown in Figure 6.
  • the crystalline form of the compound of Formula A is Form IIIA. In some embodiments, crystalline Form IIIA of the compound of Formula A is anhydrous and non-solvated. This crystalline form can be generally prepared as described in Example 4.
  • Crystalline Form IIIA of the compound of Formula A can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), and thermogravimetric analysis (TGA).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern substantially as shown in Figure 7. Peaks from the XRPD pattern are listed in Table 3.
  • crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 8.1° ⁇ 0.2°. In some embodiments, crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 10.6° ⁇ 0.2°. In some embodiments, crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 13.5° ⁇ 0.2°. In some embodiments, crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 14.2° ⁇ 0.2°. In some embodiments, crystalline Form IIIA of the compound of Formula A is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 20.3° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 2Q: 8.1° ⁇ 0.2°; 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; and 14.2° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 2Q: 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 14.2° ⁇ 0.2°; 16.4° ⁇ 0.2°; 17.1° ⁇ 0.2°; 17.9° ⁇ 0.2°; 20.3° ⁇ 0.2°; 20.8° ⁇ 0.2°; 24.1° ⁇ 0.2°; 24.6° ⁇ 0.2°; 24.8° ⁇ 0.2°; and 27.5° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising the following peaks, in terms of 2Q: 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 14.2° ⁇ 0.2°; and 20.3° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 2Q: 8.1° ⁇ 0.2°; 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 14.2° ⁇ 0.2°; 16.4° ⁇ 0.2°; 17.1° ⁇ 0.2°; 17.9° ⁇ 0.2°; 20.3° ⁇ 0.2°; and 24.1° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 14.2° ⁇ 0.2°; 16.4° ⁇ 0.2°; 17.1° ⁇ 0.2°; 17.9° ⁇ 0.2°; 20.3° ⁇ 0.2°; 20.8° ⁇ 0.2°; 24.1° ⁇ 0.2°; 24.6° ⁇ 0.2°; 24.8° ⁇ 0.2°; and 27.5° ⁇ 0.2°.
  • crystalline Form IIIA of the compound of Formula A has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 10.6° ⁇ 0.2°; 13.5° ⁇ 0.2°; 14.2° ⁇ 0.2°; 16.4° ⁇ 0.2°; 17.1° ⁇ 0.2°; 17.9° ⁇ 0.2°; 20.3° ⁇ 0.2°; and 24.1° ⁇ 0.2°.
  • Form IIIA of the compound of Formula A is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 143 °C.
  • crystalline Form IIIA of the compound of Formula A has a DSC thermogram substantially as shown in Figure 8.
  • crystalline Form IIIA of the compound of Formula A has a TGA trace substantially as shown in Figure 9.
  • the present application provides a hydrobromic acid salt of the compound of Formula A.
  • the hydrobromic acid salt of the compound of Formula A is a 1 : 1 stoichiometric ratio of 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4-methylphenyl)- 3,3,3-trifluoro-2-hydroxypropanamide to hydrobromic acid.
  • the hydrobromic acid salt form can be generally prepared as described in Example 11.
  • the hydrobromic acid salt of the compound of Formula A is crystalline. In some embodiments, the hydrobromic acid salt of the compound of Formula A is a solvated crystalline form. In some embodiments, the hydrobromic acid salt of the compound of Formula A is a methanol solvate crystalline form.
  • the compound of Formula B can be isolated in numerous crystalline forms, including, for example, crystalline forms which are anhydrous and/or non-solvated.
  • the crystalline forms of the compound of Formula B are anhydrous.
  • the crystalline forms of the compound of Formula B are non-solvated.
  • the crystalline forms of the compound of Formula B are anhydrous and non-solvated.
  • anhydrous is meant that the crystalline form of the compound of Formula B contains essentially no bound water in the crystal lattice structure, i.e.. the compound does not form a crystalline hydrate.
  • the crystalline forms provided herein can be prepared, for example, by a process comprising dissolving 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-methyl-5-(l,l,l-trifluoro-2- hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide in a solvent to form a mixture and crystallizing the compound from the mixture.
  • the process further comprises heating the mixture to a temperature of from about 70°C to about 90°C. In some embodiments, the process further comprises cooling the mixture to room temperature.
  • the solvent comprises isopropyl acetate. In some embodiments, the solvent further comprises heptane.
  • the present application provides a crystalline form of the compound 8- amino-N-(2-hydroxy-2-methylpropyl)-3-(2-methyl-5-( 1,1,1 -trifluoro-2-hydroxypropan-2- yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide, which is prepared according to a process provided herein.
  • the crystalline form is Form IB as described herein.
  • the crystalline form is Form IIB as described herein.
  • the crystalline forms of the invention are substantially isolated.
  • substantially isolated is meant that a particular crystalline form of the compound of Formula B is at least partially isolated from impurities.
  • a crystalline form of the invention comprises less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 2.5%, less than about 1%, or less than about 0.5% of impurities.
  • Impurities generally include anything that is not the substantially isolated crystalline form including, for example, other crystalline forms and other substances.
  • a crystalline form of the compound of Formula B is substantially free of other crystalline forms.
  • the phrase“substantially free of other crystalline forms” means that a particular crystalline form of the compound of Formula B comprises greater than about 80%, greater than about 90%, greater than about 95%, greater than about 98%, greater than about 99% or greater than about 99.5% by weight of the particular crystalline form.
  • the crystalline form of the compound of Formula B is Form IB. In some embodiments, crystalline Form IB of the compound of Formula B is anhydrous and non-solvated.
  • the preparation of the compound of Formula B is described in Examples 5 and 6. Crystalline Form IB can be generally prepared as described in Example 7.
  • Crystalline Form IB of the compound of Formula B can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD) and differential scanning calorimetry (DSC).
  • crystalline Form IB of the compound of Formula B is characterized by an XRPD pattern substantially as shown in Figure 10. Peaks from the XRPD pattern are listed in Table 4.
  • crystalline Form IB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 20, at 6.2° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 20, at 15.6° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 20, at 20.7° ⁇ 0.2°. In some embodiments, crystalline Form IB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.7° ⁇ 0.2°; 20.7° ⁇ 0.2°; and 23.2° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 10.4° ⁇ 0.2°; 11.4° ⁇ 0.2°; 11.6° ⁇ 0.2°; 12.0° ⁇ 0.2°; 13.9° ⁇ 0.2°; 14.4° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 20.7° ⁇ 0.2°; and 23.2° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 12.0 0 ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 19.3° ⁇ 0.2°; 20.7° ⁇ 0.2°; 23.2° ⁇ 0.2°; and 27.1° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 10.4° ⁇ 0.2°; 11.4° ⁇ 0.2°; 11.6° ⁇ 0.2°; 12.0° ⁇ 0.2°; 13.9° ⁇ 0.2°; and 14.4° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 20: 6.2° ⁇ 0.2°;
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 12.0 0 ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 19.3° ⁇ 0.2°; 20.7° ⁇ 0.2°; 23.2° ⁇ 0.2°; and 27.1° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 20.7° ⁇ 0.2°; and 23.2° ⁇ 0.2°.
  • crystalline Form IB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 10.4° ⁇ 0.2°; 11.4° ⁇
  • Form IB of the compound of Formula B is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 174 °C.
  • crystalline Form IB of the compound of Formula B has a DSC thermogram substantially as shown in Figure 11.
  • Crystalline Form IIB of the compound of Formula B is Form IIB. In some embodiments, crystalline Form IIB of the compound of Formula B is anhydrous and non-solvated. This crystalline form can be generally prepared as described in Example 8.
  • Crystalline Form IIB of the compound of Formula B can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD) and differential scanning calorimetry (DSC).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern substantially as shown in Figure 12. Peaks from the XRPD pattern are listed in Table 5.
  • crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 4.2° ⁇ 0.2°. In some embodiments, crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 7.4° ⁇ 0.2°. In some embodiments, crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 13.3° ⁇ 0.2°. In some
  • crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 20.1° ⁇ 0.2°. In some embodiments, crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 17.0°
  • crystalline Form IIB of the compound of Formula B is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 18.8° ⁇ 0.2°.
  • crystalline Form IIB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 2Q: 4.3° ⁇ 0.2°; 7.4° ⁇ 0.2°; 13.3° ⁇ 0.2°; and 15.3° ⁇ 0.2°.
  • crystalline Form IIB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 2Q: 4.3° ⁇ 0.2°; 7.4° ⁇ 0.2°; 15.3° ⁇ 0.2°; 17.0° ⁇ 0.2°; 18.8° ⁇ 0.2°; and 20.1° ⁇ 0.2°.
  • crystalline Form IIB of the compound of Formula B has an XRPD pattern comprising the following peaks, in terms of 2Q: 4.3° ⁇ 0.2°; 7.4° ⁇ 0.2°; 13.3° ⁇ 0.2°; 15.3° ⁇ 0.2°; 15.5° ⁇ 0.2°; 17.0° ⁇ 0.2°; 17.2° ⁇ 0.2°; 18.1° ⁇ 0.2°; 18.8° ⁇ 0.2°; 19.6° ⁇ 0.2°; 20.1° ⁇ 0.2°; 21.4° ⁇ 0.2°; 23.5° ⁇ 0.2°; 25.8° ⁇ 0.2°; 26.2° ⁇ 0.2°; and 27.3° ⁇ 0.2°.
  • crystalline Form IIB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 2Q: 4.3° ⁇ 0.2°; 7.4° ⁇ 0.2°; 13.3° ⁇ 0.2°; 15.3° ⁇ 0.2°; 15.5° ⁇ 0.2°; 17.0° ⁇ 0.2°; 17.2° ⁇ 0.2°; 18.1° ⁇ 0.2°; 18.8° ⁇ 0.2°; 19.6° ⁇ 0.2°; 20.1° ⁇ 0.2°; 21.4° ⁇ 0.2°; 23.5° ⁇ 0.2°; 25.8° ⁇ 0.2°; 26.2° ⁇ 0.2°; and 27.3° ⁇ 0.2°.
  • crystalline Form IIB of the compound of Formula B has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 2Q: 4.3° ⁇ 0.2°; 7.4° ⁇ 0.2°; 13.3 0 ⁇ 0.2°; 15.3° ⁇ 0.2°; 15.5° ⁇ 0.2°; 17.0° ⁇ 0.2°; 17.2° ⁇ 0.2°; 18.8° ⁇ 0.2°; and 20.1° ⁇ 0.2°.
  • Form IIB of the compound of Formula B is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 165 °C.
  • crystalline Form IIB of the compound of Formula B has a DSC thermogram substantially as shown in Figure 13.
  • the compound of Formula C can be isolated in numerous crystalline forms, including, for example, crystalline forms which are anhydrous and/or non-solvated.
  • the crystalline forms of the compound of Formula C are anhydrous.
  • the crystalline forms of the compound of Formula C are non-solvated.
  • the crystalline forms of the compound of Formula C are anhydrous and non-solvated.
  • anhydrous is meant that the crystalline form of the compound of Formula C contains essentially no bound water in the crystal lattice structure, i.e.. the compound does not form a crystalline hydrate.
  • the present application provides a process of preparing a crystalline form of the compound 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-(methyl-d 3 )-5-(l, l,l-trifluoro-2- hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide.
  • the process comprises dissolving 8-amino-N-(2-hydroxy-2-methylpropyl)-3-(2-(methyl-d 3 )-5-(l,l,l-trifluoro-2- hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide in a solvent to form a mixture and crystallizing the compound from the mixture.
  • the process further comprises heating the mixture to a temperature of from about 70°C to about 90°C. In some embodiments, the process further comprises cooling the mixture to room temperature.
  • the solvent comprises isopropyl acetate. In some embodiments, the solvent further comprises heptane.
  • the present application further provides a crystalline form of the compound 8-amino-N-(2- hydroxy-2-methylpropyl)-3 -(2-(methyl-d 3 )-5 -( 1 , 1 , 1 -trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo [ 1 ,2- a]pyrazine-6-carboxamide, which is prepared according a process provided herein.
  • the crystalline form is Form IC as described herein.
  • the crystalline forms of the invention are substantially isolated.
  • substantially isolated is meant that a particular crystalline form of the compound of Formula C is at least partially isolated from impurities.
  • a crystalline form of the invention comprises less than about 50%, less than about 40%, less than about 30%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 2.5%, less than about 1%, or less than about 0.5% of impurities.
  • Impurities generally include anything that is not the substantially isolated crystalline form including, for example, other crystalline forms and other substances.
  • a crystalline form of the compound of Formula C is substantially free of other crystalline forms.
  • phrases“substantially free of other crystalline forms” means that a particular crystalline form of the compound of Formula C comprises greater than about 80%, greater than about 90%, greater than about 95%, greater than about 98%, greater than about 99% or greater than about 99.5% by weight of the particular crystalline form.
  • the crystalline form of the compound of Formula C is crystalline Form IC. In some embodiments, crystalline Form IC of the compound of Formula C is anhydrous and non-solvated.
  • the preparation of the compound of Formula C of the compound of Formula C is described in Example 9. Crystalline Form IC of the compound of Formula C can be generally prepared as described in Example 10
  • Crystalline Form IC of the compound of Formula C can be identified by unique signatures with respect to, for example, X-ray powder diffraction (XRPD) and differential scanning calorimetry (DSC).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • crystalline Form IC of the compound of Formula C is characterized by an XRPD pattern substantially as shown in Figure 14. Peaks from the XRPD pattern are listed in Table 6.
  • crystalline Form IC of the compound of Formula C is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 6.2° ⁇ 0.2°. In some embodiments, crystalline Form IC of the compound of Formula C is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 11.9° ⁇ 0.2°. In some embodiments, crystalline Form IC of the compound of Formula C is characterized by an XRPD pattern comprising a peak, in terms of 2Q, at 16.7° ⁇ 0.2°.
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising the following peaks, in terms of 2Q: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 18.8° ⁇ 0.2°; 19.9° ⁇ 0.2°; 20.7° ⁇ 0.2°; 21.2° ⁇ 0.2°; 22.3° ⁇ 0.2°; 23.2° ⁇ 0.2°; and 27.0 0 ⁇ 0.2°.
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising the following peaks, in terms of 2Q: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; and 21.2° ⁇ 0.2°.
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising the following peaks, in terms of 2Q: 6.2° ⁇ 0.2°; 10.4° ⁇ 0.2°; 11.3° ⁇ 0.2°; 11.9° ⁇ 0.2°; and 12.5° ⁇ 0.2°
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 2Q: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 18.8° ⁇ 0.2°; 19.9° ⁇ 0.2°; 20.7° ⁇ 0.2°; 21.2° ⁇ 0.2°; 22.3° ⁇ 0.2°; 23.2° ⁇ 0.2°; and 27.0 0 ⁇ 0.2°.
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 2Q: 6.2° ⁇ 0.2°; 10.4° ⁇ 0.2°; 11.3° ⁇ 0.2°; 11.9° ⁇ 0.2°; 12.5° ⁇ 0.2°; 13.8° ⁇ 0.2°; 14.4° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 20.7° ⁇ 0.2°; and 21.2° ⁇ 0.2°.
  • crystalline Form IC of the compound of Formula C has an XRPD pattern comprising 2 or more, 3 or more, or 4 or more of the following peaks, in terms of 20: 6.2° ⁇ 0.2°; 15.6° ⁇ 0.2°; 16.0° ⁇ 0.2°; 16.7° ⁇ 0.2°; 20.7° ⁇ 0.2°; and 21.2° ⁇ 0.2°.
  • Form IC of the compound of Formula C is characterized by a DSC thermogram comprising an endothermic peak having a maximum at about 179 °C.
  • crystalline Form IC of the compound of Formula C has a DSC thermogram substantially as shown in Figure 15.
  • the compounds (e.g., salts and crystalline forms) of the invention described herein inhibit activity of RI3Kg kinase. Accordingly, the salts and crystalline forms of the invention described herein can be used in methods of inhibiting RI3Kg kinase by contacting the kinase. In some embodiments, the salts and crystalline forms of the invention can be used in methods of inhibiting activity of RI3Kg in an individual/patient in need of the inhibition by administering an effective amount of a salt or crystalline form described herein. In some embodiments, modulating is inhibiting. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is ex vivo.
  • the crystalline forms as described herein demonstrate better efficacy and favorable safety and toxicity profiles in animal studies.
  • the RI3Kg includes a mutation.
  • a mutation can be a replacement of one amino acid for another, or a deletion of one or more amino acids.
  • the mutation can be present in the kinase domain of the RI3Kg.
  • the salt or crystalline form further inhibits PI3K5.
  • the salts and crystalline forms described herein can be selective.
  • selective is meant that the salt or crystalline form binds to or inhibits RI3Kg with greater affinity or potency, respectively, compared to at least one other kinase.
  • the salts and crystalline forms of the disclosure are selective inhibitors of RI3Kg over PI3K5, PI3Ka, and RI3Kb.
  • the salts and crystalline forms of the disclosure are selective inhibitors of RI3Kg over PI3Ka and RI3Kb.
  • selectivity can be at least about 2-fold, 3-fold, 5-fold, lO-fold, at or 20-fold over PI3K5 as measured by the assays described herein. In some embodiments, selectivity can be tested at the 2 mM ATP concentration of each enzyme. In some embodiments, the selectivity of salts and crystalline forms of the disclosure can be determined by cellular assays associated with particular PI3K kinase activity.
  • Another aspect of the present disclosure pertains to methods of treating a kinase RBKg-associated disease or disorder in an individual (e.g. , patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of one or more salts or crystalline forms of the present disclosure or a pharmaceutical composition thereof.
  • a RBKg-associated disease or disorder can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the RI3Kg, including overexpression and/or abnormal activity levels.
  • the disease or disorder is an autoimmune disease or disorder, cancer, cardiovascular disease, or neurodegenerative disease.
  • the disease or disorder is lung cancer (e.g., non-small cell lung cancer), melanoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, color cancer, endometrial cancer, bladder cancer, skin cancer, cancer of the uterus, renal cancer, gastric cancer, or sarcoma.
  • lung cancer e.g., non-small cell lung cancer
  • melanoma pancreatic cancer
  • breast cancer prostate cancer
  • liver cancer color cancer
  • endometrial cancer bladder cancer
  • skin cancer cancer of the uterus
  • renal cancer gastric cancer
  • sarcoma sarcoma
  • the sarcoma is Askin's tumor, sarcoma botryoides, chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, alveolar soft part sarcoma, angiosarcoma, cystosarcoma phyllodes, dermatofibrosarcoma protuberans, desmoid tumor, desmoplastic small round cell tumor, epithelioid sarcoma, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, fibrosarcoma, gastrointestinal stromal tumor (GIST), hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant peripheral nerve sheath tumor (MPNST), neurofibrosarcom
  • the disease or disorder is mesothelioma or adrenocarcinoma. In some embodiments, the disease or disorder is mesothelioma. In some embodiments, the disease or disorder is adrenocarcinoma.
  • the disease or disorder is acute myeloid leukemia (e.g., acute monocytic leukemia), small lymphocyctic lymphoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), multiple myeloma, T-cell actute lymphoblasic leukemia (T-ALL), cutaneous T-cell lymphoma, large granular lymphocytic leukemia, mature (peripheral) t-cell neoplasm (PTCL), anaplastic large cell lymphoma (ALCL), or lymphoblastic lymphoma.
  • acute myeloid leukemia e.g., acute monocytic leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • T-ALL T-cell actute lymphoblasic leukemia
  • cutaneous T-cell lymphoma large granular lymphocytic leukemia
  • PTCL mature (
  • t-cell neoplasm is T-cell prolymphocytic leukemia, T-cell granular lymphocytic leukemia, aggressive NK-cell leukemia, mycosis fimgoides/Sezary syndrome, naplastic large cell lymphoma (T-cell type), enteropathy type T-cell lymphoma, adult T-cell leukemia/lymphoma, or angioimmunoblastic T-cell lymphoma
  • the anaplastic large cell lymphoma is systemic ALCL or primary cutaneous ALCL.
  • the disease or disorder is Burkitt's lymphoma, acute myeloblastic leukemia, chronic myeloid leukemia, non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, hairy cell leukemia, Mantle cell lymphoma, small lymphocytic lymphoma, follicular lymphoma, xenoderoma pigmentosum, keratoctanthoma, lymphoplasmacytic lymphoma, extranodal marginal zone lymphoma, Waldenstrom's macroglobulinemia, prolymphocytic leukemia, acute lymphoblastic leukemia, myelofibrosis, mucosa-associated lymphatic tissue (MALT) lymphoma, mediastinal (thymic) large B-cell lymphoma, lymphomatoid granulomatosis, splenic marginal zone lymphoma, primary effusion lymphoma, intravascular large B-cell lymphoma,
  • MALT
  • MDSC myeloid-derived suppressor cells
  • myeloid lineage a family of cells that originate from bone marrow stem cells.
  • MDSCs strongly expand in pathological situations such as chronic infections and cancer, as a result of an altered haematopoiesis.
  • MDSCs are discriminated from other myeloid cell types in which they possess strong immunosuppressive activities rather than immunostimulatory properties. Similar to other myeloid cells, MDSCs interact with other immune cell types including T cells, dendritic cells, macrophages and natural killer cells to regulate their functions.
  • the compounds, etc. described herein can be used in methods related to cancer tissue (e.g., tumors) with high infiltration of MDSCs, including Solid tumors with high basal level of macrophage and/or MDSC infiltration.
  • the disease or disorder is Burkitf s lymphoma, acute myeloblastic leukemia, chronic myeloid leukemia, non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, hairy cell leukemia, Mantle cell lymphoma, small lymphocytic lymphoma, follicular lymphoma,
  • lymphoplasmacytic lymphoma extranodal marginal zone lymphoma, Waldenstrom's macroglobulinemia, prolymphocytic leukemia, acute lymphoblastic leukemia, myelofibrosis, mucosa-associated lymphatic tissue (MALT) lymphoma, mediastinal (thymic) large B-cell lymphoma, lymphomatoid granulomatosis, splenic marginal zone lymphoma, primary effusion lymphoma, intravascular large B-cell lymphoma, plasma cell leukemia, extramedullary plasmacytoma, smouldering myeloma (aka asymptomatic myeloma), monoclonal gammopathy of undetermined significance (MGUS), or diffuse large B cell lymphoma.
  • MALT mucosa-associated lymphatic tissue
  • MALT mucosa-associated lymphatic tissue lymphoma
  • MALT lymphoma mediastinal (thymic) large B-cell
  • the non-Hodgkin’s lymphoma is relapsed NHL, refractory NHL, recucurrent follicular NHL, indolent NHL (iNHL), or aggressive NHL (aNHL).
  • the diffuse large B cell lymphoma is activated B-cell like (ABC) diffuse large B cell lymphoma, or germinal center B cell (GCB) diffuse large B cell lymphoma.
  • ABSC activated B-cell like
  • GCB germinal center B cell
  • the Burkitf s lymphoma is endemic Burkitf s lymphoma, sporadic Burkitf s lymphoma, or Burkitf s-like lymphoma.
  • the disease or disorder is rheumatoid arthritis, multiple sclerosis, systemic lupus erythematous, asthma, allergy (e.g, allergic rhinitis), pancreatitis, psoriasis, anaphylaxis, glomerulonephritis, inflammatory bowel disease (e.g., Crohn’s disease and ulcerative colitis), thrombosis, meningitis, encephalitis, diabetic retinopathy, benign prostatic hypertrophy, myasthenia gravis, Sjogren’s syndrome, osteoarthritis, restenosis, or atherosclerosis.
  • allergy e.g, allergic rhinitis
  • pancreatitis psoriasis
  • anaphylaxis glomerulonephritis
  • glomerulonephritis glomerulonephritis
  • inflammatory bowel disease e.g., Crohn’s disease and ulcerative colitis
  • thrombosis meningit
  • the disease or disorder is heart hypertropy, cardiac myocyte dysfunction, acute coronary syndrome, chronic obstructive pulmonary disease (COPD), chronic bronchitis, elevated blood pressure, ischemia, ischemia-reperfusion, vasoconstriction, anemia (e.g., hemolytic anemia, aplastic anemia, or pure red cell anemia), bacterial infection, viral infection, graft rejection, kidney disease, anaphylactic shock fibrosis, skeletal muscle atrophy, skeletal muscle hypertrophy, angiogenesis, sepsis, graft-versus-host disease, allogeneic or xenogeneic transplantation, glomerulosclerosis, progressive renal fibrosis, idiopathic thrombocytopenic purpura (ITP), idiopathic pulmonary fibrosis, autoimmune hemolytic anemia, vasculitis, lupus nephritis, pemphigus, or membranous nephropathy.
  • COPD chronic obstructive
  • disease or disorder is heart hypertropy, cardiac myocyte dysfunction, chronic obstructive pulmonary disease (COPD), elevated blood pressure, ischemia, ischemia-reperfusion, vasoconstriction, anemia (e.g., hemolytic anemia, aplastic anemia, or pure red cell anemia), bacterial infection, viral infection, graft rejection, kidney disease, anaphylactic shock fibrosis, skeletal muscle atrophy, skeletal muscle hypertrophy, angiogenesis, sepsis, graft rejection, glomerulosclerosis, progressive renal fibrosis, idiopathic thrombocytopenic purpura (ITP), autoimmune hemolytic anemia, vasculitis, systemic lupus erythematosus, lupus nephritis, pemphigus, or membranous nephropathy.
  • COPD chronic obstructive pulmonary disease
  • anemia e.g., hemolytic anemia, aplastic anemia, or pure red cell an
  • the disease or disorder is Alzheimer's disease, central nervous system trauma, or stroke.
  • the idiopathic thrombocytopenic purpura is relapsed ITP or refractory ITP.
  • the vasculitis is Behcet's disease, Cogan's syndrome, giant cell arteritis, polymyalgia rheumatica (PMR), Takayasu's arteritis, Buerger's disease (thromboangiitis obliterans), central nervous system vasculitis, Kawasaki disease, polyarteritis nodosa, Churg-Strauss syndrome, mixed cryoglobulinemia vasculitis (essential or hepatitis C virus (HCV)-induced), Henoch-Schonlein purpura (HSP), hypersensitivity vasculitis, microscopic polyangiitis, Wegener's granulomatosis, or anti neutrophil cytoplasm antibody associated (ANCA) systemic vasculitis (AASV).
  • ANCA neutrophil cytoplasm antibody associated
  • the present disclosure further provides a salt or crystalline form described herein, for use in any of the methods described herein.
  • the present disclosure further provides use of a crystalline form described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in any of the methods described herein.
  • “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • “contacting” a PI3K with a salt or crystalline form of the disclosure includes the administration of a crystalline form of the present disclosure to an individual or patient, such as a human, having a PI3K, as well as, for example, introducing a salt or crystalline form of the disclosure into a sample containing a cellular or purified preparation containing the PI3K.
  • the salts and crystalline forms provided herein, or any of the embodiments thereof may possess satisfactory pharmacological profile and promising biopharmaceutical properties, such as toxicological profile, metabolism and pharmacokinetic properties, solubility, and permeability. It will be understood that determination of appropriate biopharmaceutical properties is within the
  • the term“individual” or“patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the phrase“therapeutically effective amount” refers to the amount of active salt, crystalline form, or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • the term“treating” or“treatment” can refer to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or
  • symptomatology such as decreasing the severity of disease.
  • the salts and crystalline forms of the invention are useful in preventing or reducing the risk of developing any of the diseases referred to herein; e.g., preventing or reducing the risk of developing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease.
  • the RI3Kg inhibitors provided herein can be used in combination with one or more immune checkpoint inhibitors for the treatment of cancer as described herein. In one
  • the combination with one or more immune checkpoint inhibitors as described herein can be used for the treatment of melanoma.
  • Salts and crystalline forms of the present disclosure can be used in combination with one or more immune checkpoint inhibitors.
  • Exemplary immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CD20, CD28, CD40, CD 122, CD96, CD73, CD47, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, HPK1, CD137 (also known as 4-1BB), ICOS, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, TIGIT, PD-l, PD-L1 and PD-L2.
  • the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, 0X40, GITR and CD137.
  • the immune checkpoint molecule is an inhibitory checkpoint molecule selected from A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-l, TIM3, TIGIT, and VISTA.
  • the salts and crystalline forms of the disclosure provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD 160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.
  • the RI3Kg inhibitors provided herein can be used in combination with one or more agonists of immune checkpoint molecules, e.g., 0X40, CD27, 0X40, GITR, and CD 137 (also known as 4-1BB).
  • immune checkpoint molecules e.g., 0X40, CD27, 0X40, GITR, and CD 137 (also known as 4-1BB).
  • the inhibitor of an immune checkpoint molecule is anti-PD 1 antibody, anti -PD-L 1 antibody, or anti-CTLA-4 antibody.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-l, e.g., an anti-PD-l monoclonal antibody.
  • the anti-PD-l monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), durvalumab (Imfinzi®), pidilizumab, SHR-1210, PDR001, MGA012, PDR001, AB122, or AMP -224.
  • the anti-PD-l monoclonal antibody is nivolumab or pembrolizumab.
  • the anti-PD 1 antibody is
  • the anti-PD-l monoclonal antibody is MGA012. In some embodiments, the anti-PD 1 antibody is SHR-1210.
  • Other anti-cancer agent(s) include antibody therapeutics such as 4-1BB (e.g. urelumab, utomilumab).
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L 1, e.g., an anti-PD-Ll monoclonal antibody.
  • the anti-PD-Ll monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), or MSB0010718C.
  • the anti-PD-Ll monoclonal antibody is MPDL3280A or MEDI4736.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-l and PD-L1, e.g., an anti-PD- 1/PD-L1 monoclonal antibody.
  • the anti-PD- 1/PD-L1 is MCLA-136.
  • the inhibitor is MCLA-145.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA- 4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab, tremelimumab, AGEN1884, or CP-675,206.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody.
  • the anti-LAG3 antibody is BMS-986016, LAG525, or INCAGN2385.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of TIM3, e.g., an anti-TIM3 antibody.
  • the anti-TIM3 antibody is INCAGN2390, MBG453, or TSR-022.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody.
  • the anti-GITR antibody is TRX518, MK-4166, INCAGN1876, MK-1248, AMG228, BMS-986156, GWN323, or MEDI1873.
  • the inhibitor of an immune checkpoint molecule is an agonist of 0X40, e.g., 0X40 agonist antibody or OX40L fusion protein.
  • the anti-OX40 antibody is MEDI0562, MOXR-0916, PF-04518600, GSK3174998, or BMS-986178.
  • the OX40L fusion protein is MEDI6383.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CD20, e.g., an anti-CD20 antibody.
  • the anti-CD20 antibody is obinutuzumab or rituximab.
  • the salts and crystalline forms of the present disclosure can be used in combination with bispecific antibodies.
  • one of the domains of the bispecific antibody targets PD-l, PD-L1, CTLA-4, GITR, 0X40, TIM3, LAG3, CD137, ICOS, CD3 or TGFp receptor.
  • the RI3Kg inhibitors provided herein can be used in combination with one or more metabolic enzyme inhibitors.
  • the metabolic enzyme inhibitor is an inhibitor of IDOl, TDO, or arginase.
  • IDOl inhibitors include epacadostat, NLG919, BMS- 986205, PF-06840003, IOM2983, RG-70099 and LY338196.
  • the additional compounds, inhibitors, agents, etc. can be combined with the present compound in a single or continuous dosage form, or they can be administered simultaneously or sequentially as separate dosage forms.
  • Cancer cell growth and survival can be impacted by multiple signaling pathways.
  • Targeting more than one signaling pathway (or more than one biological molecule involved in a given signaling pathway) may reduce the likelihood of drug-resistance arising in a cell population, and/or reduce the toxicity of treatment.
  • the salts and crystalline forms of the present disclosure can be used in combination with one or more other enzyme/protein/receptor inhibitors or one or more therapies for the treatment of diseases, such as cancer.
  • diseases and indications treatable with combination therapies include those as described herein.
  • cancers include solid tumors and liquid tumors, such as blood cancers.
  • One or more additional pharmaceutical agents such as, for example, chemotherapeutics, anti inflammatory agents, steroids, immunosuppressants, immune -oncology agents, metabolic enzyme inhibitors, chemokine receptor inhibitors, and phosphatase inhibitors, as well as targeted therapies such as Bcr-Abl, Flt-3, EGFR, HER2, JAK, c-MET, VEGFR, PDGFR, c-Kit, IGF-1R, RAF and FAR kinase inhibitors such as, for example, those described in WO 2006/056399.
  • Other agents such as therapeutic antibodies can be used in combination with the salts and crystalline forms of the present disclosure for treatment of PI3K-associated diseases, disorders or conditions.
  • the one or more additional pharmaceutical agents such as, for example, chemotherapeutics, anti inflammatory agents, steroids, immunosuppressants, immune -oncology agents, metabolic enzyme inhibitors, chemokine receptor inhibitors, and phosphatase inhibitors, as well as targeted therapies such
  • pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • the salts and crystalline forms as disclosed herein can be combined with one or more inhibitors of the following kinases for the treatment of cancer and other diseases or disorders described herein: Aktl, Akt2, Akt3, TGF-pR, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IGF-1R, IR-R, PDGFaR, PDGFpR, CSFIR, KIT, FFK-II, KDR/FFK-l, FFK-4, flt-l, FGFR1, FGFR2, FGFR3, FGFR4, c-Met, Ron, Sea, TRKA, TRKB, TRKC, FFT3, VEGFR/Flt2, Flt4, EphAl, EphA2, EphA3, EphB2, EphB4, Tie2, Src,
  • Non-limiting examples of inhibitors that can be combined with the salts and crystalline forms of the present disclosure for treatment of cancer and other diseases and disorders described herein include an FGFR inhibitor (FGFR1, FGFR2, FGFR3 or FGFR4, e.g., INCB54828, INCB62079 and INCB63904), a JAK inhibitor (JAK1 and/or JAK2, e.g., ruxolitinib, baricitinib or INCB39110), an IDO inhibitor (e.g., epacadostat, NFG919, or BMS-986205), an FSD1 inhibitor (e.g., INCB59872 and INCB60003), a TDO inhibitor, a PI3K-delta inhibitor (e.g., INCB50797 and INCB50465), a Pim inhibitor,
  • FGFR inhibitor FGFR1, FGFR2, FGFR3 or FGFR4, e.g., INCB54828, INCB62079 and INCB
  • the salts and crystalline forms described herein are administered with a PI3K5 inhibitor. In some embodiments, the salts and crystalline forms described herein are administered with a JAK inhibitor. In some embodiments, the salts and crystalline forms described herein are administered with a JAK1 or JAK2 inhibitor (e.g., baricitinib or ruxolitinib). In some embodiments, the salts and crystalline forms described herein are administered with a JAK1 inhibitor. In some
  • the salts and crystalline forms described herein are administered with a JAK1 inhibitor, which is selective over JAK2.
  • Example antibodies for use in combination therapy include but are not limited to Trastuzumab (e.g. anti-HER2), Ranibizumab (e.g. anti-VEGF-A), Bevacizumab (trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGFR), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
  • Trastuzumab e.g. anti-HER2
  • Ranibizumab e.g. anti-VEGF-A
  • Bevacizumab trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGFR), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
  • agents may be used in combination with the salts and crystalline forms of the present disclosure and are presented as a non-limiting list: a cytostatic agent, cisplatin, doxorubicin, taxotere, taxol, etoposide, irinotecan, camptostar, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methoxtrexate, temozolomide, cyclophosphamide, SCH 66336,
  • the salts and crystalline forms of the present disclosure can further be used in combination with other methods of treating cancers, for example by chemotherapy, irradiation therapy, tumortargeted therapy, adjuvant therapy, immunotherapy or surgery.
  • immunotherapy include cytokine treatment (e.g., interferons, GM-CSF, G-CSF, IL-2), CRS-207 immunotherapy, cancer vaccine, monoclonal antibody, adoptive T cell transfer, Toll receptor agonists, STING agonists, oncolytic virotherapy and immunomodulating small molecules, including thalidomide or JAK1/2 inhibitor and the like.
  • the salts and crystalline forms can be administered in combination with one or more anti-cancer drugs, such as a chemotherapeutics.
  • Example chemotherapeutics include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, baricitinib, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxe
  • chemotherapeutics include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • proteosome inhibitors e.g., bortezomib
  • thalidomide thalidomide
  • revlimid thalidomide
  • DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include corticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include imatinib mesylate (GLEEVACTM), nilotinib, dasatinib, bosutinib, and ponatinib, and pharmaceutically acceptable salts.
  • Other example suitable Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184, WO 04/005281, and U.S. Ser. No. 60/578,491.
  • Example suitable Flt-3 inhibitors include midostaurin, lestaurtinib, linifanib, sunitinib, sunitinib, maleate, sorafenib, quizartinib, crenolanib, pacritinib, tandutinib, PLX3397 and ASP2215, and their pharmaceutically acceptable salts.
  • Other example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120.
  • Example suitable RAF inhibitors include dabrafenib, sorafenib, and vemurafenib, and their pharmaceutically acceptable salts.
  • Other example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
  • Example suitable FAK inhibitors include VS-4718, VS-5095, VS-6062, VS-6063, BI853520, and GSK2256098,and their pharmaceutically acceptable salts.
  • Other example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.
  • the salts and crystalline forms of the disclosure can be used in
  • kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • the salts and crystalline forms of the disclosure can be used in
  • the salts and crystalline forms of the disclosure can be used in combination with a chemotherapeutic provided herein.
  • additional pharmaceutical agents used in the treatment of multiple myeloma can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib).
  • Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors.
  • the agent is an alkylating agent, a proteasome inhibitor, a corticosteroid, or an
  • an alkylating agent examples include cyclophosphamide (CY), melphalan (MEF), and bendamustine.
  • the proteasome inhibitor is carfdzomib.
  • the corticosteroid is dexamethasone (DEX).
  • the immunomodulatory agent is lenalidomide (FEN) or pomalidomide (POM). Additive or synergistic effects are desirable outcomes of combining a PI3K inhibitor of the present disclosure with an additional agent.
  • the salts and crystalline forms of the disclosure can be used in
  • the agents can be combined with the present compound in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • the salts and crystalline forms of the present disclosure can be used in combination with one or more other inhibitors or one or more therapies for the treatment of infections.
  • infections include viral infections, bacterial infections, fungus infections or parasite infections.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with the compounds of the disclosure where the dexamethasone is administered
  • the salts and crystalline forms of the present disclosure as described herein can be combined with another immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • tumor vaccines include peptides of melanoma antigens, such as peptides of gplOO, MAGE antigens, Trp-2, MARTI and/or tyrosinase, or tumor cells transfected to express the cytokine GM-CSF.
  • the salts and crystalline forms of the present disclosure as described herein can be used in combination with a vaccination protocol for the treatment of cancer.
  • the tumor cells are transduced to express GM-CSF.
  • tumor vaccines include the proteins from viruses implicated in human cancers such as Human Papilloma Viruses (HPV), Hepatitis Viruses (HBV and HCV) and Kaposi's Herpes Sarcoma Virus (KHSV).
  • the salts and crystalline forms of the present disclosure can be used in combination with tumor specific antigen such as heat shock proteins isolated from tumor tissue itself.
  • the salts and crystalline forms can be combined with dendritic cells immunization to activate potent anti -tumor responses.
  • the salts and crystalline forms of the present disclosure can be used in combination with bispecific macrocyclic peptides that target Fe alpha or Fe gamma receptor-expressing effectors cells to tumor cells.
  • the salts and crystalline forms of the present disclosure can also be combined with macrocyclic peptides that activate host immune responsiveness.
  • combinations of the salts and crystalline forms of the disclosure with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
  • the salts and crystalline forms of the present disclosure can be used in combination with bone marrow transplant for the treatment of a variety of tumors of hematopoietic origin.
  • the salts and crystalline forms can be used in combination with vaccines, to stimulate the immune response to pathogens, toxins, and self antigens.
  • pathogens for which this therapeutic approach may be particularly useful, include pathogens for which there is currently no effective vaccine, or pathogens for which conventional vaccines are less than completely effective. These include, but are not limited to, HIV, Hepatitis (A, B, & C), Influenza, Herpes, Giardia, Malaria,
  • Viruses causing infections treatable by methods of the present disclosure include, but are not limit to human papillomavirus, influenza, hepatitis A, B, C or D viruses, adenovirus, poxvirus, herpes simplex viruses, human cytomegalovirus, severe acute respiratory syndrome virus, ebola virus, measles virus, herpes virus (e.g., VZV, HSV-l, HAV-6, HSV-II, and CMV, Epstein Barr virus), flaviviruses, echovirus, rhinovirus, coxsackie virus, comovirus, respiratory syncytial virus, mumpsvirus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus.
  • human papillomavirus influenza, hepatitis
  • Pathogenic bacteria causing infections treatable by methods of the disclosure include, but are not limited to, chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lyme's disease bacteria.
  • Pathogenic fungi causing infections treatable by methods of the disclosure include, but are not limited to, Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
  • Candida albicans, krusei, glabrata, tropicalis, etc.
  • Cryptococcus neoformans Aspergillus (fumigatus, niger, etc.)
  • Genus Mucorales micor, absidia, rhizophus
  • Sporothrix schenkii Blastomyces dermatitidis
  • Paracoccidioides brasiliensis C
  • Pathogenic parasites causing infections treatable by methods of the disclosure include, but are not limited to, Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia,
  • Cryptosporidium sp. Pneumocystis carinii
  • Plasmodium vivax Babesia microti
  • Trypanosoma brucei Trypanosoma cruzi
  • Leishmania donovani Toxoplasma gondi
  • Nippostrongylus brasiliensis
  • the compounds (e.g., salts and crystalline forms) of the disclosure can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • topical including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery
  • pulmonary e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, the compound of the disclosure or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • the compounds of the disclosure may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • Finely divided (nanoparticulate) preparations of the compounds of the disclosure can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the disclosure can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1000 mg (1 g), more usually about 100 to about 500 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • compositions of the disclosure contain from about 5 to about 50 mg of the active ingredient.
  • the compositions of the disclosure contain from about 5 to about 50 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compositions containing about 5 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, about 25 to about 30, about 30 to about 35, about 35 to about 40, about 40 to about 45, or about 45 to about 50 mg of the active ingredient.
  • compositions of the disclosure contain from about 50 to about 500 mg of the active ingredient.
  • compositions of the disclosure contain from about 500 to about 1000 mg of the active ingredient.
  • the active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present disclosure.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present disclosure.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present disclosure.
  • the tablets or pills of the present disclosure can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present disclosure can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • Carrier selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the disclosure.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of a compound of the present disclosure can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the disclosure in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the compounds of the disclosure can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 pg/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the disclosure can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • labeled compounds e.g., salts and crystalline forms
  • the disclosure radio-labeled, fluorescent-labeled, etc.
  • assays both in vitro and in vivo, for localizing and quantitating PI3K in tissue samples, including human, and for identifying PI3K ligands by inhibition binding of a labeled compound.
  • Substitution of one or more of the atoms of the compounds of the present disclosure can also be useful in generating differentiated ADME (Adsorption, Distribution, Metabolism and Excretion.)
  • ADME Adsorption, Distribution, Metabolism and Excretion.
  • the present disclosure includes PI3K assays that contain such labeled or substituted compounds.
  • the present disclosure further includes isotopically-labeled compounds of the disclosure.
  • An “isotopically” or“radio-labeled” compound is a compound of the disclosure where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e.. naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present disclosure include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, 35 S, 36 Cl, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 I, 125 I and 13 1 1.
  • one or more hydrogen atoms in a compound of the present disclosure can be replaced by deuterium atoms (e.g., one or more hydrogen atoms of a Ci- 6 alkyl group of Formula (I) can be optionally substituted with deuterium atoms, such as -CD 3 being substituted for -CH 3 ).
  • alkyl groups of the disclosed Formulas and/or Forms can be perdeute rated.
  • the compound includes at least one deuterium atom.
  • one or more hydrogen atoms in a compound presented herein can be replaced or substituted by deuterium (e.g., one or more hydrogen atoms of a C 1-6 alkyl group can be replaced by deuterium atoms, such as -CD 3 being substituted for -CFF).
  • the compound includes two or more deuterium atoms.
  • the compound includes 1, 1-2, 1-3, 1-4, 1-5, or 1-6 deuterium atoms.
  • all of the hydrogen atoms in a compound can be replaced or substituted by deuterium atoms.
  • 1, 2, 3, 4, 5, 6, 7, or 8 hydrogen atoms, attached to carbon atoms of any Formula A substituents are each optionally replaced by a deuterium atom.
  • Isotopically labeled compounds can used in various studies such as NMR spectroscopy, metabolism experiments, and/or assays.
  • substitution with heavier isotopes may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances (see e.g. , A. Kerekes et.al. J. Med. Chem. 2011, 54, 201-210; R. Xu et.al. J. Label Compd. Radiopharm. 2015, 58, 308-312).
  • substitution at one or more metabolism sites may afford one or more of the therapeutic advantages.
  • radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro PI3K labeling and competition assays, compounds that incorporate 3 H, 14 C, 82 Br, 125 I, 131 I or 35 S can be useful. For radio imaging applications U C, 18 F, 125 I, 123 I, 124 I, 131 1, 75 Br, 76 Br or 77 Br can be useful.
  • a“radio-labeled” or“labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 1, 35 S and 82 Br.
  • the present disclosure can further include synthetic methods for incorporating radio-isotopes into compounds of the disclosure. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of disclosure.
  • a labeled compound of the disclosure can be used in a screening assay to identify /evaluate compounds.
  • a newly synthesized or identified compound i.e. , test compound
  • a test compound which is labeled can be evaluated for its ability to bind a PI3K by monitoring its concentration variation when contacting with the PI3K, through tracking of the labeling.
  • a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a PI3K (i.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to the PI3K directly correlates to its binding affinity.
  • the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.
  • kits useful for example, in the treatment or prevention of PI3K-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a salts and crystalline forms of the disclosure.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • Preparatory LC-MS purifications of some of the compounds prepared were performed on Waters mass directed fractionation systems.
  • the basic equipment setup, protocols, and control software for the operation of these systems have been described in detail in the literature (see e.g.,“Two-Pump At Column Dilution Configuration for Preparative LC-MS”, K. Blom, J. Combi. Chem., 4, 295 (2002);“Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis Purification”, K. Blom, R. Sparks, J. Doughty, G. Everlof, T. Haque, A. Combs, J. Combi.
  • pH 2 purifications: Waters SunfireTM Cis 5 pm, 30 x 100 mm or Waters XBridgeTM Cis 5 pm,
  • pH 10 purifications: Waters XBridgeTM Cis 5 pm, 30 x 100 mm column, eluting with mobile phase A: 0.1% NH 4 OH in water and mobile phase B: acetonitrile; the flow rate was 60 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature (see e.g. "Preparative LCMS Purification: Improved Compound Specific Method Optimization", K. Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004)).
  • X-Ray Powder Diffraction analysis was carried out on a Bruker D8 Advance ECO X-ray Powder Diffractometer (XRPD) instrument with the following parameters: radiation source is Cu at 1.5418 A and LYNXEYETM detector and X-ray power of 40 KV, 25 mA.
  • the sample powder was dispersed on a zero-background sample holder.
  • General measurement conditions were: Start Angle - 3°; Stop Angle - 30°; Sampling - 0.015 deg.; and Scan speed - 2 deg/min.
  • DSC Differential Scanning Calorimetry
  • Thermogravimetric analysis was carried out on a TA Instrument Thermogravimetric Analyzer, TGA5500 with an autosampler at the following conditions: Ramp at 10 °C/min. from 25 °C to 600 °C; nitrogen gas at 25 mL/min balance purge flow; and platinum sample pan.
  • Step 1 1 -(3-Bromo-4-methylphenyl)-2,2,2-trifluoroethan-l -ol
  • reaction mixture was cooled back to 0 °C and treated with water (6.48 mL, 360 mmol) and 1.0 M tetrabutylammonium fluoride in tetrahydrofuran (6.54 mL, 6.54 mmol). The ice bath was removed and the reaction mixture was stirred at ambient temperature for 30 min. The yellow reaction mixture was diluted with brine (150 mL) and extracted with ethyl acetate (200 mL). The organic layer was washed with saturated ammonium chloride (100 mL), dried over sodium sulfate, filtered, and concentrated to give a tan oil.
  • the reaction mixture was concentrated (by rotary evaporation with the water bath set at 28 °C) to give a rust colored solid.
  • the solid was dissolved in THF (29.6 mL), cooled to 0 °C, treated with 1.8 M HC1 (10.9 mL, 19.6 mmol), and stirred at room temperature (rt) for 1.5 h.
  • the reaction mixture was diluted with water (75 mL) and extracted with diethyl ether (3 x 75 mL). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated. Reconcentration from hexanes gave the desired product (8.70 g, 99.8%) as an orange solid that was used without further purification.
  • LCMS for CgLLBrFsO (M- CN) + : m/z 267.0, 269.0; Found: 266.9, 269.0.
  • the reaction mixture was bubbled with nitrogen for 10 min to remove some of the HC1, cooled to 0 °C, and diluted with brine (200 mL), water (50 mL), and ethyl acetate (200 mL).
  • the organic layer was separated and the aqueous layer was diluted with water (100 mL) to dissolve the remaining solids.
  • the aqueous layer was extracted with ethyl acetate (100 mL).
  • the combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated to give a brown oil. Purification by flash column
  • the reaction mixture was treated with ethyl acetate (200 mL) and brine (50 mL), filtered over Celite and the Celite was rinsed with additional ethyl acetate.
  • the aqueous layer from the filtrate was separated and extracted with ethyl acetate (200 mL).
  • the combined organic extracts were dried over sodium sulfate, filtered, and concentrated to a brown foam. Purification by flash column chromatography using MeOH in dichloromethane (0% to 10%) gave the desired product as a red/brown foam that was not completely pure.
  • This material was repurified by flash column chromatography using MeOH in dichloromethane (0% to 15%) to give the desired product as an orange/brown foam that was still not completely pure.
  • This material was repurified by flash column chromatography using ethyl acetate (containing 5% MeOH) in hexanes (0% to 100%) to give the desired product as a white foam that still contained an impurity.
  • This material was repurified by flash column chromatography using acetonitrile (containing 5% MeOH) in dichloromethane (0% to 100%) to give the desired product (4.67 g, 40.4%) as a white foam.
  • a vial was charged with 2-(3-(8-amino-6-(trifluoromethyl)imidazo[l,2-a]pyrazin-3-yl)-4- methylphenyl)-3,3,3-trifluoro-2-hydroxypropanamide (0.050 g, 0.115 mmol) and stirred at 80 °C while a 1:2 premixed solution of isopropyl acetate (0.676 mL) / heptane (1.34 mL) was added dropwise. After 2 mL was added the solid was not completely dissolved and some remained on the bottom of the vial. After almost all of the solids had dissolved, new solids were forming on the walls of the vial.
  • Form IA was confirmed as a crystalline solid according to XRPD analysis.
  • the XRPD pattern of Form IA is shown in Figure 1 and the peak data is given below in Table 1.
  • TGA analysis of Form IA revealed significant weight loss above 200 °C due to decomposition of the sample .
  • the TGA thermogram is provided in Figure 3.
  • Form IA was confirmed as an anhydrous, non-solvated crystalline form.
  • Form IIA was confirmed as a crystalline solid according to XRPD analysis.
  • the XRPD pattern of Form IIA is shown in Figure 4 and the peak data is given below in Table 2.
  • DSC analysis of Form IIA revealed one endothermic peak with an onset temperature of 177.2 °C and a maximum at 179.7 °C.
  • the DSC thermogram is provided in Figure 5.
  • TGA analysis of Form IIA revealed significant weight loss above 200 °C due to decomposition of the sample.
  • the TGA thermogram is provided in Figure 6.
  • Form IIIA was confirmed as a crystalline solid according to XRPD analysis.
  • Form IIIA is shown in Figure 7 and the peak data is given below in Table 3.
  • TGA analysis of Form IIIA revealed significant weight loss above 200 °C due to decomposition of the sample.
  • the TGA thermogram is provided in Figure 9.
  • Form IIIA was confirmed as an anhydrous, non-solvated crystalline form.
  • Step 5 lJ, l-Trifluoro-2-(4-methyl-3-(4,4,5,5-tetramethyl-J 3,2-dioxaborolan-2-yl)phenyl)propan-2-ol
  • tetrahydrofuran (4.95 mL) was degassed with nitrogen for 5 min.
  • the reaction mixture was treated with triphenylphosphine palladium chloride (0.025 g, 0.036 mmol), degassed with nitrogen for another 5 min, and heated at 135 °C in the microwave for 20 min.
  • the reaction mixture was diluted with ethyl acetate and fdtered through a 0.5 micrometer cartridge that was rinsed with ethyl acetate.
  • the fdtrate was washed with water and brine, dried over sodium sulfate, fdtered, and concentrated to give a crude residue.
  • the reaction mixture was concentrated and the resultant red oil was diluted with ethyl acetate, water, and saturated sodium bicarbonate.
  • the aqueous layer was separated and re-extracted with ethyl acetate (2x).
  • the combined organic layers were washed with brine, dried over magnesium sulfate, filtered, and
  • reaction mixture was treated with additonal trimethylaluminum (0.70 ml, 1.40 mmol) (2 M in toluene) and stirred at 80 °C overnight.
  • the reaction mixture was cooled to room temperature, diluted with methanol, and filtered over a pad of Celite®. After rinsing with MeOH (2x), the filtrate was concentrated to an amber oil. Purification via silica gel chromatography (0-5% MeOH/DCM) afforded the title compound as an oily solid (26 mg,
  • the racemic mixture was separated via preparative chiral HPLC (Phenomenex Lux Amylose-l [2l.2x250mm, 5 micron], eluting with 12% ethanol in hexanes, at flow rate of 18 mL/min, loading ⁇ 8 mg in 800 pL ethanol).
  • the first peak that eluted had a retention time of 11.9 min (Example 1; Enantiomer 1).
  • the second peak that eluted had a retention time of 16.1 min (Example 2, Enantiomer 2).
  • Example 7 Preparation and Characterization of 8-Amino-N-(2-hydroxy-2-methylpropyl)-3-(2- methyl-5-(l,l,l-trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide, Crystalline Form IB (Free Base)
  • Form IB was confirmed as a crystalline solid according to XRPD analysis.
  • the XRPD pattern of Form IB is shown in Figure 10 and the peak data is given below in Table 4.
  • Form IB DSC analysis of Form IB revealed one endothermic peak with an onset temperature of 172.2 °C and a maximum at 174.2 °C.
  • the DSC thermogram is provided in Figure 11.
  • Form IB was confirmed as an anhydrous, non-solvated crystalline form.
  • Example 8 Preparation and Characterization of 8-Amino-N-(2-hydroxy-2-methylpropyl)-3-(2- methyl-5-(l,l,l-trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6-carboxamide, Crystalline Form IIB (Free Base)
  • Form IIB was confirmed as a crystalline solid according to XRPD analysis.
  • the XRPD pattern of Form IIB is shown in Figure 12 and the peak data is given below in Table 5.
  • Form IIB DSC analysis of Form IIB revealed one endothermic peak with an onset temperature of 161.7 °C and a maximum at 165.4 °C.
  • the DSC thermogram is provided in Figure 8.
  • Form IIB was confirmed as an anhydrous, non-solvated crystalline form.
  • Step 1 1 -(4-ftiethyl-ch)phenyl)ethan-l -one
  • dichloromethane/HCl/ice mixture with additional dichloromethane The mixture was warmed to room temperature (rt) and the layers were separated. The aqueous layer was extracted with dichloromethane (2 x 75 mL). The combined organic layers were washed with saturated sodium bicarbonate and brine. The sodium bicarbonate and brine washes contained product and these were combined, acidified with 1M HC1, and extracted with dichloromethane (2 x 50 mL). The organic layers were all combined, dried over magnesium sulfate, filtered, and concentrated to a yellow oil. Purification by flash column
  • tetrahydrofuran (168 mL) at 0 °C was treated with trimethyl(trifluoromethyl)silane (8.07 mL, 58.8 mmol) [Aldrich, 488712] and stirred at 0 °C for 5 min.
  • the reaction mixture was treated with 1.0 M tetra-n- butylammonium fluoride in tetrahydrofuran (2.10 mL, 2.10 mmol) at 0 °C and stirred at room temperature for 1 h.
  • the reaction mixture was treated with 1.0 M tetra-n-butylammonium fluoride in tetrahydrofuran (12.6 mL, 12.6 mmol) and water (9.8 mL) and stirred at room temperature for 30 min.
  • the reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered, and concentrated to give a crude residue. Purification by flash column chromatography using ethyl acetate in hexanes (0% - 20%) gave the desired product (13.3 g, 111%) as a yellow oil.
  • Step 4. 1 ,7, l-Trifluoro-2-(4-(methyl-d 3 )-3-(4,4,5,5-tetramethyl-J3,2-dioxaborolan-2-yl)phenyl)propan-2- ol
  • reaction mixture was treated with bis(triphenylphosphine)palladium(II) dichloride (1.16 g, 1.67 mmol), degassed with nitrogen for another 10 min, and stirred at 100 °C overnight.
  • the reaction mixture was cooled to rt, degassed with nitrogen for 5 min, treated with additional
  • the reaction mixture was treated with 1.0 M potassium carbonate in water (107 ml, 107 mmol), degassed with nitrogen for 5 min, and stirred at 80 °C overnight.
  • the reaction mixture was cooled to rt and filtered over Celite®.
  • the Celite® was rinsed with ethyl acetate and water.
  • the filtrate was diluted with water (150 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated to a dark oil.
  • Step 6 Second eluting enantiomer of 2-(3-(8-amino-6-bromoimidazo[l,2-a]pyrazin-3-yl)-4-(methyl- dfphenyl)-!, 1 J-trifluoropropan-2-ol
  • the racemic mixture of 2-(3-(8-amino-6-bromoimidazo[l,2-a]pyrazin-3-yl)-4-(methyl- d 3 )phenyl)-l,l,l-trifluoropropan-2-ol was separated via preparative chiral HPLC (Phenomenex Lux Amylose-l [2l.2x250mm, 5 micron], eluting with 20% ethanol in hexanes, at flow rate of 20 mL/min, loading ⁇ 200 mg in 4 mL ethanol).
  • the first peak that eluted had a retention time of 9.6 min.
  • the second peak that eluted had a retention time of 14.6 min.
  • Step 7 Methyl 8-amino-3-(2-(methyl-ds)-5-(l, l,l-trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[l, 2- a]pyrazine-6-carboxylate (single enantiomer prepared)
  • the reaction mixture was treated with PdidppfkOLCL (0.998 g, 1.22 mmol), degassed with nitrogen for another 5 min, saturated with CO by bubbling the gas through the reaction subsurface for 3 min, and heated at 60 °C overnight.
  • the reaction mixture was concentrated and the resultant oil was diluted with ethyl acetate and water..
  • the aqueous layer was separated and re-extracted with ethyl acetate (3x).
  • the combined organic layers were washed with water, saturated ammonium chloride solution, and brine, dried over magnesium sulfate, filtered, and concentrated to a brown oil.
  • the combined ethyl acetate layers were extracted with additional 1.0 M sodium hydroxide (3 x 20 mL).
  • the combined basic aqueous layers were adjusted to pH ⁇ 5 with citric acid (7.6 g).
  • the aqueous layer was extracted with dichloromethane (2 x 150 mL).
  • the aqueous layer was diluted with brine and extracted with ethyl acetate (150 mL).
  • the combined organic layers were concentrated to give the desired product (4.06 g, 93.8%) as atan solid that was used without further purification.
  • Step 9 8-Amino-N-(2-hydroxy-2-methylpropyl)-3-(2-(methyl-ds)-5-(J Jl-trifluoro-2-hydroxypropan-2- yl)phenyl)imidazo[J 2-a]pyrazine-6-carboxamide
  • reaction mixture was diluted with water (500 mL) and brine (100 mL) and extracted with ethyl acetate (3 x 150 mL). The combined organics were washed with saturated ammonium chloride (150 mL), 11% sodium carbonate (150 mL), and brine (100 mL), dried over magnesium sulfate, filtered, and concentrated to an amber oil. Purification by flash column
  • Example 10 Preparation and Characterization of 8-Amino-N-(2-hydroxy-2-methylpropyl)-3-(2- (methyl-d3)-5-(l,l,l-trifluoro-2-hydroxypropan-2-yl)phenyl)imidazo[l,2-a]pyrazine-6- carboxamide, Crystalline Form IC (Free Base)
  • Form IC was confirmed as a crystalline solid according to XRPD analysis.
  • the XRPD pattern of Form IC is shown in Figure 14 and the peak data is given below in Table 6.
  • Form IC was confirmed as an anhydrous, non-solvated crystalline form.
  • Table 7 shows atomic coordinates (xlO 4 ) and equivalent isotropic displacement parameters (A 2 x 10 3 ).
  • U(eq) is defined as one third of the trace of the orthogonalized Uij tensor.
  • Table 8 shows bond lengths [A] and angles [deg].
  • Table 9 shows anisotropic displacement parameters (A 2 x 10 3 ).
  • THP-l cells Human Acute Monocytic Leukemia
  • ATCC Manassas, VA
  • FBS Fluorescence-Activated Cell Sorting Factor
  • THP-l cells are serum starved overnight in RPMI, then plated in RPMI (2xl0 5 cells/well in 90 pL) into 96-well flat-bottom tissue culture treated plates (Coming, Coming, NY), in the presence or absence of a concentration range of test compounds.
  • the lysates are tested in the Human/Mouse/Rat Phospho-RPS6 EFISA (R&D Systems, Inc. Minn, MN).
  • the plate is measured using a microplate reader (SpectraMax M5 - Molecular Devices, EEC Sunnyvale, CA) set to 450 nm with a wavelength correction of 540.
  • IC50 determination is performed by fitting the curve of inhibitor percent inhibition versus the log of the inhibitor concentration using the GraphPad Prism 5.0 software.
  • [g- 33 R]ATR (10 mCi/mF) and Wheat Germ Agglutinin (WGA) YSi SPA Scintillation Beads was purchased from Perkin-Elmer (Waltham, MA).
  • Lipid kinase substrate, D-myo-Phosphatidylinositol 4,5- bisphosphate (PtdIns(4,5)P2)D (+)-sn-l,2-di-0-octanoylglyceryl, 3-O-phospho linked (PIP2), CAS 204858-53-7 was purchased from Echelon Biosciences (Salt Lake City, UT).
  • RI3Kg pl 10g
  • Recombinant Human Protein was purchased from Life technology (Grand Island, NY ).
  • ATP, MgCL, DTT, EDTA, MOPS and CHAPS were purchased from SigmaAldrich (St. Louis, MO).
  • the kinase reaction was conducted in polystyrene 384-well Greiner Bio-one white plate from Thermo Fisher Scientific in a final volume of 25 pL. Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 2%.
  • the RI3Kg assay was carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM MgCL, 5 mM DTT and CHAPS 0.03%.
  • [g- 33 R]ATR (10 mCi/mL) and Wheat Germ Agglutinin (WGA) YSi SPA Scintillation Beads was purchased from Perkin-Elmer (Waltham, MA).
  • Lipid kinase substrate, D-myo-Phosphatidylinositol 4,5- bisphosphate (PtdIns(4,5)P2)D (+)-sn-l,2-di-0-octanoylglyceryl, 3-O-phospho linked (PIP2), CAS 204858-53-7 was purchased from Echelon Biosciences (Salt Lake City, UT).
  • PI3K5 (pl 10d /p85a) Recombinant Human Protein was purchased from Eurofms (St Charles, MO). ATP, MgCh, DTT, EDTA, MOPS and CHAPS were purchased from SigmaAldrich (St. Louis, MO).
  • the kinase reaction was conducted in polystyrene 384-well Greiner Bio-one white plate from Thermo Lisher Scientific in a final volume of 25 pL. Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 2%.
  • the PI3K5 assay was carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM Mg Cl 2, 5 mM DTT and CHAPS 0.03%. Reactions were initiated by the addition of ATP, the final reaction mixture consisted of 20 mM PIP2, 2 mM ATP, 0.5 pCi [g- 33 R] ATP, 3.4 nM PI3K5.
  • + refers to IC50 of ⁇ 100 nM
  • ++ refers to IC50 of ⁇ 500 nM
  • +++ refers to an IC50 of ⁇ 2000 nM
  • ++++ refers to an IC50 of > 2000 nM.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
PCT/US2019/049419 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor WO2020051169A1 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
RS20240361A RS65334B1 (sr) 2018-09-05 2019-09-04 Kristalni oblici inhibitora fosfatidilinozitol 3-kinaze (pi3k)
AU2019336675A AU2019336675A1 (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (PI3K) inhibitor
MX2021002551A MX2021002551A (es) 2018-09-05 2019-09-04 Formas cristalinas de inhibidor de fosfoinositida 3-cinasa (pi3k).
FIEP19858443.5T FI3847175T3 (fi) 2018-09-05 2019-09-04 Fosfoinositidi-3-kinaasin (pi3k) inhibiittorin kiteisiä muotoja
CA3111629A CA3111629A1 (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
BR112021004094-3A BR112021004094A2 (pt) 2018-09-05 2019-09-04 formas cristalinas de um inibidor de fosfoinositídeo 3-quinase (pi3k)
SI201930734T SI3847175T1 (sl) 2018-09-05 2019-09-04 Kristalinične oblike zaviralca fosfoinozitid 3-kinaze (pi3k)
EP19858443.5A EP3847175B1 (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
PE2021000289A PE20211815A1 (es) 2018-09-05 2019-09-04 Formas cristalinas de un inhibidor de fosfoinositida 3-quinasa (pi3k)
EA202190679A EA202190679A1 (ru) 2018-10-17 2019-09-04 Кристаллические формы ингибитора фосфоинозитид 3-киназы (pi3k)
JP2021512576A JP2021535182A (ja) 2018-09-05 2019-09-04 ホスホイノシチド3−キナーゼ(pi3k)阻害剤の結晶形態
CR20210165A CR20210165A (es) 2018-09-05 2019-09-04 Formas cristalinas de un inhibidor de fosfoinositida 3-quinasa (pi3k) campo técnico
EP24153948.5A EP4338801A3 (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
CN201980071241.6A CN113677683A (zh) 2018-09-05 2019-09-04 磷酸肌醇3激酶(pi3k)抑制剂的结晶形式
IL309869A IL309869A (en) 2018-09-05 2019-09-04 Crystal forms of phosphoinositide 3-kinase inhibitor (PI3K)
SG11202102224UA SG11202102224UA (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
KR1020217009964A KR20210091121A (ko) 2018-09-05 2019-09-04 포스포이노시티드 3-키나아제 (pi3k) 억제제의 결정질 형태
HRP20240252TT HRP20240252T1 (hr) 2018-09-05 2019-09-04 Kristalni oblici inhibitora fosfoinozitid 3 kinaze (pi3k)
LTEPPCT/US2019/049419T LT3847175T (lt) 2018-09-05 2019-09-04 Fosfoinozitido 3-kinazės (pi3k) inhibitoriaus kristalinės formos
DK19858443.5T DK3847175T3 (da) 2018-09-05 2019-09-04 Krystallinske former af en phosphoinositid-3-kinase- (pi3k) inhibitor
IL281262A IL281262A (en) 2018-09-05 2021-03-04 Crystal forms of phosphoinositide 3-kinase inhibitor (P13K)
PH12021550472A PH12021550472A1 (en) 2018-09-05 2021-03-05 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
CONC2021/0004061A CO2021004061A2 (es) 2018-09-05 2021-03-31 Formas cristalinas de un inhibidor de fosfoinositida 3–quinasa (pi3k)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201862727339P 2018-09-05 2018-09-05
US201862727321P 2018-09-05 2018-09-05
US201862727328P 2018-09-05 2018-09-05
US62/727,328 2018-09-05
US62/727,321 2018-09-05
US62/727,339 2018-09-05
US201862746928P 2018-10-17 2018-10-17
US62/746,928 2018-10-17

Publications (1)

Publication Number Publication Date
WO2020051169A1 true WO2020051169A1 (en) 2020-03-12

Family

ID=69642051

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/049419 WO2020051169A1 (en) 2018-09-05 2019-09-04 Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor

Country Status (27)

Country Link
US (2) US11186580B2 (es)
EP (2) EP3847175B1 (es)
JP (1) JP2021535182A (es)
KR (1) KR20210091121A (es)
CN (1) CN113677683A (es)
AU (1) AU2019336675A1 (es)
BR (1) BR112021004094A2 (es)
CA (1) CA3111629A1 (es)
CL (1) CL2021000539A1 (es)
CO (1) CO2021004061A2 (es)
CR (1) CR20210165A (es)
DK (1) DK3847175T3 (es)
EC (1) ECSP21022719A (es)
FI (1) FI3847175T3 (es)
HR (1) HRP20240252T1 (es)
IL (2) IL309869A (es)
LT (1) LT3847175T (es)
MA (1) MA53561A (es)
MX (2) MX2021002551A (es)
PE (1) PE20211815A1 (es)
PH (1) PH12021550472A1 (es)
PT (1) PT3847175T (es)
RS (1) RS65334B1 (es)
SG (1) SG11202102224UA (es)
SI (1) SI3847175T1 (es)
TW (1) TWI833796B (es)
WO (1) WO2020051169A1 (es)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019079469A1 (en) 2017-10-18 2019-04-25 Incyte Corporation CONDENSED IMIDAZOLE DERIVATIVES SUBSTITUTED WITH HYDROXY TERTIARY GROUPS AS INHIBITORS OF PI3K-GAMMA

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070099925A1 (en) * 2005-09-02 2007-05-03 Calderwood David J Novel imidazo based heterocycles
US20120329792A1 (en) * 2010-03-09 2012-12-27 Janssen Pharmaceutica Nv IMIDAZO[1,2-a]PYRAZINE DERIVATIVES AND THEIR USE FOR THE PREVENTION OR TREATMENT OF NEUROLOGICAL, PSYCHIATRIC AND METABOLIC DISORDERS AND DISEASES
US20140249132A1 (en) * 2013-03-01 2014-09-04 Incyte Corporation Use of pyrazolopyrimidine derivatives for the treatment of pi3k-delta related disorders
US20150005309A1 (en) * 2012-01-10 2015-01-01 Bayer Intellectual Property Gmbh Substituted imidazopyrazines as akt kinase inhibitors
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS

Family Cites Families (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4269846A (en) 1979-10-29 1981-05-26 Usv Pharmaceutical Corporation Heterocyclic compounds useful as anti-allergy agents
US5137876A (en) 1990-10-12 1992-08-11 Merck & Co., Inc. Nucleoside antiviral and anti-inflammatory compounds and compositions and methods for using same
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
AU5620299A (en) 1998-08-11 2000-03-06 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
GB9905075D0 (en) 1999-03-06 1999-04-28 Zeneca Ltd Chemical compounds
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
GB0011092D0 (en) 2000-05-08 2000-06-28 Black James Foundation Gastrin and cholecystokinin receptor ligands (III)
CA2413330A1 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
IL160915A0 (en) 2001-09-19 2004-08-31 Aventis Pharma Sa Indolizines inhibiting kinase proteins
FR2831536A1 (fr) 2001-10-26 2003-05-02 Aventis Pharma Sa Nouveaux derives de benzimidazoles, leur procede de preparation, leur application a titre de medicament, compositions pharmaceutiques et nouvelle utilisation notamment comme inhibiteurs de kdr
WO2003035065A1 (en) 2001-10-26 2003-05-01 Aventis Pharmaceuticals Inc Benzimidazoles and analogues and their use as protein kinases inhibitors
TWI302836B (en) 2001-10-30 2008-11-11 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
DE10207843A1 (de) 2002-02-15 2003-09-04 Schering Ag Mikrolia-Inhibitoren zur Unterbrechung von Interleukin 12 und IFN-gamma vermittelten Immunreaktionen
TW200406374A (en) 2002-05-29 2004-05-01 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
TWI335913B (en) 2002-11-15 2011-01-11 Vertex Pharma Diaminotriazoles useful as inhibitors of protein kinases
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
US7186832B2 (en) 2003-02-20 2007-03-06 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
US7157460B2 (en) 2003-02-20 2007-01-02 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
WO2004078943A2 (en) 2003-03-04 2004-09-16 California Institute Of Technology Alternative heterocycles for dna recognition
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
BRPI0413234A (pt) 2003-08-01 2006-10-03 Genelabs Tech Inc derivados de imidazola bicìclica contra flaviviridae
AR045944A1 (es) 2003-09-24 2005-11-16 Novartis Ag Derivados de isoquinolina 1.4-disustituidas
WO2005118580A2 (en) 2004-05-12 2005-12-15 The Government Of The United States Of America As Represented By The Secretary, Department Of Health Tricyclic compounds as inhibitors of the hypoxic signaling pathway
MXPA06014247A (es) 2004-06-10 2007-03-12 Irm Llc Compuestos y composiciones como inhibidores de la proteina quinasa.
CN101106983A (zh) 2004-11-24 2008-01-16 诺瓦提斯公司 JAK抑制剂与至少一种Bcr-Abl、Flt-3、FAK或RAF激酶抑制剂的组合
WO2007019346A1 (en) 2005-08-04 2007-02-15 Sirtris Pharmaceuticals, Inc. Benzothiazoles and thiazolopyridines as sirtuin modulators
ES2340093T3 (es) 2006-06-22 2010-05-28 Biovitrum Ab (Publ) Derivados pirazina y piridina como inhibidores de cinasa mnk.
CA2681813A1 (en) 2007-03-27 2009-01-08 Paratek Pharmaceuticals, Inc. Transcription factor modulating compounds and methods of use thereof
DE102007035333A1 (de) 2007-07-27 2009-01-29 Boehringer Ingelheim Pharma Gmbh & Co. Kg Neue substituierte Arylsulfonylglycine, deren Herstellung und deren Verwendung als Arzneimittel
GB0716292D0 (en) 2007-08-21 2007-09-26 Biofocus Dpi Ltd Imidazopyrazine compounds
AU2008321046B2 (en) 2007-11-16 2013-10-24 Incyte Holdings Corporation 4-pyrazolyl-N-arylpyrimidin-2-amines and 4-pyrazolyl-N-heteroarylpyrimidin-2-amines as janus kinase inhibitors
EP2234487A4 (en) 2007-12-19 2011-09-07 Scripps Research Inst ANILIDES AND ANALOGUES AS INHIBITORS OF RHO KINASE
CA2712071A1 (en) 2008-01-15 2009-10-08 Siga Technologies, Inc. Antiviral drugs for treatment of arenavirus infection
ATE555116T1 (de) 2008-02-26 2012-05-15 Merck Sharp & Dohme Ahcy-hydrolasehemmer zur behandlung von hyperhomocysteinämie
SG191660A1 (en) 2008-03-11 2013-07-31 Incyte Corp Azetidine and cyclobutane derivatives as jak inhibitors
WO2009133127A1 (en) 2008-04-30 2009-11-05 Merck Serono S.A. Fused bicyclic compounds and use thereof as pi3k inhibitors
WO2009158118A2 (en) 2008-05-30 2009-12-30 University Of Notre Dame Du Lac Anti-bacterial agents from benzo[d]heterocyclic scaffolds for prevention and treatment of multidrug resistant bacteria
US8470819B2 (en) 2008-11-03 2013-06-25 Merck Sharp & Dohme Corp. Benzimidazole and aza-benzimidazole carboxamides
WO2010061903A1 (ja) 2008-11-27 2010-06-03 塩野義製薬株式会社 Pi3k阻害活性を有するピリミジン誘導体およびピリジン誘導体
GB0822981D0 (en) 2008-12-17 2009-01-21 Summit Corp Plc Compounds for treatment of duchenne muscular dystrophy
EP2401273A1 (en) 2009-02-27 2012-01-04 Vertex Pharmaceuticals Incorporated Tri-cyclic pyrazolopyridine kinase inhibitors
EA025520B1 (ru) 2009-05-22 2017-01-30 Инсайт Холдингс Корпорейшн N-(ГЕТЕРО)АРИЛПИРРОЛИДИНОВЫЕ ПРОИЗВОДНЫЕ ПИРАЗОЛ-4-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ И ПИРРОЛ-3-ИЛ-ПИРРОЛО[2,3-d]ПИРИМИДИНОВ В КАЧЕСТВЕ ИНГИБИТОРОВ ЯНУС-КИНАЗЫ
CN104945420A (zh) 2009-06-29 2015-09-30 因塞特公司 作为pi3k抑制剂的嘧啶酮类
TW201113285A (en) 2009-09-01 2011-04-16 Incyte Corp Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US8759359B2 (en) 2009-12-18 2014-06-24 Incyte Corporation Substituted heteroaryl fused derivatives as PI3K inhibitors
TW201130842A (en) 2009-12-18 2011-09-16 Incyte Corp Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
WO2011099832A2 (en) 2010-02-12 2011-08-18 Crystalgenomics, Inc. Novel benzimidazole compound, preparation method thereof and pharmaceutical composition comprising the same
NZ602313A (en) 2010-03-10 2014-08-29 Incyte Corp Piperidin-4-yl azetidine derivatives as jak1 inhibitors
JP2013523766A (ja) 2010-03-31 2013-06-17 グラクソ グループ リミテッド キナーゼ阻害剤としてのイミダゾリル‐イミダゾール
JP5816678B2 (ja) 2010-04-14 2015-11-18 インサイト・コーポレイションIncyte Corporation PI3Kδ阻害剤としての縮合誘導体
EP2575866A4 (en) 2010-05-24 2013-10-16 Presidio Pharmaceuticals Inc HCV NS5A INHIBITORS
US8623857B2 (en) 2010-05-26 2014-01-07 Merck Sharp & Dohme Corp. N-phenyl imidazole carboxamide inhibitors of 3-phosphoinositide-dependent protein kinase-1
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
AR083450A1 (es) 2010-10-13 2013-02-27 Millennium Pharm Inc Heteroarilos y sus usos en el tratamiento de enfermedades proliferativas, inflamatorias o cardiovasculares
CA2818542A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
WO2012068440A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
EP2649062B1 (en) 2010-11-30 2015-04-08 Takeda Pharmaceutical Company Limited Bicyclic compounds as inhibitors of acetyl-coa carboxylase (acc)
AR084366A1 (es) 2010-12-20 2013-05-08 Incyte Corp N-(1-(fenil sustituido)etil)-9h-purin-6-aminas como inhibidores de pi3k
WO2012125629A1 (en) 2011-03-14 2012-09-20 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as pi3k inhibitors
US8673905B2 (en) 2011-03-17 2014-03-18 Hoffmann-La Roche Inc. Imidazo pyrazines
WO2012135009A1 (en) 2011-03-25 2012-10-04 Incyte Corporation Pyrimidine-4,6-diamine derivatives as pi3k inhibitors
KR20140093610A (ko) 2011-04-21 2014-07-28 재단법인 한국파스퇴르연구소 소염 화합물
EA031618B1 (ru) 2011-06-09 2019-01-31 Ризен Фармасьютикалз Са Соединения-модуляторы gpr-119
WO2012177606A1 (en) 2011-06-20 2012-12-27 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
TW201313721A (zh) 2011-08-18 2013-04-01 Incyte Corp 作為jak抑制劑之環己基氮雜環丁烷衍生物
HUE043703T2 (hu) 2011-09-02 2019-09-30 Incyte Holdings Corp Heterociklusos aminok PI3K inhibitorokként
WO2013129674A1 (ja) 2012-03-01 2013-09-06 学校法人兵庫医科大学 新規ベンズイミダゾール誘導体及びその用途
AR090548A1 (es) 2012-04-02 2014-11-19 Incyte Corp Azaheterociclobencilaminas biciclicas como inhibidores de pi3k
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
BR112014029501B1 (pt) 2012-05-31 2020-05-19 Sumitomo Chemical Co composto heterocíclico fundido, seu uso, agente de controle de pragas e método para o controle de pragas
CA2877550C (en) 2012-07-13 2020-09-08 Ucb Biopharma Sprl Imidazopyridine derivatives as modulators of tnf activity
CN104918945B (zh) 2012-11-01 2018-01-05 因赛特公司 作为jak抑制剂的三环稠合噻吩衍生物
WO2014149207A2 (en) 2013-03-15 2014-09-25 Dow Agrosciences Llc Benzimidazole-based insecticidal compositions and related methods
US8999992B2 (en) 2013-03-15 2015-04-07 Vm Pharma Llc Crystalline forms of tryosine kinase inhibitors and their salts
WO2014153529A1 (en) 2013-03-22 2014-09-25 The Scripps Research Institute Substituted benzimidazoles as nociceptin receptor modulators
EP2994142A4 (en) 2013-05-08 2017-03-29 Colorado Seminary, Which Owns and Operates The University of Denver Antibiotic and anti-parasitic agents that modulate class ii fructose 1,6-bisphosphate aldolase
SI2997023T1 (sl) 2013-05-17 2017-07-31 Incyte Corporation Bipirazolni derivati kot inhibitorji JAK
SG10201800325PA (en) 2013-07-17 2018-02-27 Otsuka Pharma Co Ltd Cyanotriazole compounds
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
ES2829914T3 (es) 2014-04-08 2021-06-02 Incyte Corp Tratamiento de enfermedades malignas de células B mediante una combinación de inhibidor de JAK y PI3K
EP2930048A1 (en) 2014-04-10 2015-10-14 Johnson Controls Automotive Electronics SAS Head up display projecting visual information onto a screen
US10077277B2 (en) 2014-06-11 2018-09-18 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US9676770B2 (en) 2014-09-16 2017-06-13 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10023576B2 (en) * 2014-10-22 2018-07-17 Bristol-Myers Squibb Company Heteroaryl substituted pyrrolotriazine amine compounds as PI3K inhibitors
EP3209664B1 (en) 2014-10-22 2020-06-03 Bristol-Myers Squibb Company Bicyclic heteroaryl amine compounds as pi3k inhibitors
MA40933A (fr) 2014-11-11 2017-09-19 Piqur Therapeutics Ag Difluorométhyl-aminopyridines et difluorométhyl-aminopyrimidines
WO2016130501A1 (en) 2015-02-09 2016-08-18 Incyte Corporation Aza-heteroaryl compounds as pi3k-gamma inhibitors
US9968604B2 (en) 2015-04-16 2018-05-15 Chiesi Farmaceutici S.P.A. Chromene derivatives as phoshoinositide 3-kinases inhibitors
WO2017079519A1 (en) 2015-11-06 2017-05-11 Incyte Corporation Heterocyclic compounds as pi3k-gamma inhibitors
AR107293A1 (es) 2016-01-05 2018-04-18 Incyte Corp COMPUESTOS DE PIRIDINA Y PIRIDIMINA COMO INHIBIDORES DE PI3K-g
WO2018038265A1 (ja) 2016-08-26 2018-03-01 田辺三菱製薬株式会社 二環式含窒素複素環化合物
WO2019079469A1 (en) * 2017-10-18 2019-04-25 Incyte Corporation CONDENSED IMIDAZOLE DERIVATIVES SUBSTITUTED WITH HYDROXY TERTIARY GROUPS AS INHIBITORS OF PI3K-GAMMA

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070099925A1 (en) * 2005-09-02 2007-05-03 Calderwood David J Novel imidazo based heterocycles
US20120329792A1 (en) * 2010-03-09 2012-12-27 Janssen Pharmaceutica Nv IMIDAZO[1,2-a]PYRAZINE DERIVATIVES AND THEIR USE FOR THE PREVENTION OR TREATMENT OF NEUROLOGICAL, PSYCHIATRIC AND METABOLIC DISORDERS AND DISEASES
US20150005309A1 (en) * 2012-01-10 2015-01-01 Bayer Intellectual Property Gmbh Substituted imidazopyrazines as akt kinase inhibitors
US20140249132A1 (en) * 2013-03-01 2014-09-04 Incyte Corporation Use of pyrazolopyrimidine derivatives for the treatment of pi3k-delta related disorders
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Physicians' Desk Reference", 1996, MEDICAL ECONOMICS COMPANY
CHEM, vol. 54, 2011, pages 201 - 210
R. XU, J. LABEL COMPD. RADIOPHARM, vol. 58, 2015, pages 308 - 312
See also references of EP3847175A4

Also Published As

Publication number Publication date
BR112021004094A2 (pt) 2021-05-25
PT3847175T (pt) 2024-04-16
CN113677683A (zh) 2021-11-19
EP4338801A2 (en) 2024-03-20
EP3847175A1 (en) 2021-07-14
TW202024088A (zh) 2020-07-01
KR20210091121A (ko) 2021-07-21
IL309869A (en) 2024-02-01
AU2019336675A1 (en) 2021-03-25
LT3847175T (lt) 2024-03-25
DK3847175T3 (da) 2024-03-18
MX2023013908A (es) 2023-12-08
HRP20240252T1 (hr) 2024-05-24
EP3847175B1 (en) 2024-02-07
CO2021004061A2 (es) 2021-06-10
EP3847175A4 (en) 2022-05-11
CA3111629A1 (en) 2020-03-12
SG11202102224UA (en) 2021-04-29
PH12021550472A1 (en) 2021-11-22
US11186580B2 (en) 2021-11-30
US20220112198A1 (en) 2022-04-14
JP2021535182A (ja) 2021-12-16
ECSP21022719A (es) 2021-06-30
MA53561A (fr) 2022-05-11
IL281262A (en) 2021-04-29
EP4338801A3 (en) 2024-06-05
CL2021000539A1 (es) 2021-08-20
RS65334B1 (sr) 2024-04-30
FI3847175T3 (fi) 2024-03-22
TWI833796B (zh) 2024-03-01
MX2021002551A (es) 2021-07-15
CR20210165A (es) 2021-10-01
PE20211815A1 (es) 2021-09-14
US20200071335A1 (en) 2020-03-05
SI3847175T1 (sl) 2024-05-31

Similar Documents

Publication Publication Date Title
EP3762368B1 (en) Aminopyrazine diol compounds as pi3k-y inhibitors
US11926630B2 (en) Tertiary alcohols as PI3K-γ inhibitors
US11046658B2 (en) Aminopyrazine derivatives as PI3K-γ inhibitors
US11161838B2 (en) Heterocyclic derivatives as PI3K inhibitors
WO2020102216A1 (en) Substituted heterocyclic derivatives as pi3k inhibitors
WO2019126505A1 (en) 3-(5-amino-pyrazin-2-yl)-benzenesulfonamide derivatives and related compounds as pi3k-gamma kinase inhibitors for treating e.g. cancer
US20220112198A1 (en) Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
EA044976B1 (ru) Кристаллические формы ингибитора фосфоинозитид 3-киназы (pi3k)
WO2021158891A1 (en) Salts and solid forms and processes of preparing a pi3k inhibitor
EA043981B1 (ru) ТРЕТИЧНЫЕ СПИРТЫ В КАЧЕСТВЕ ИНГИБИТОРОВ PI3K-γ

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19858443

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3111629

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021512576

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2101001279

Country of ref document: TH

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021004094

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019336675

Country of ref document: AU

Date of ref document: 20190904

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019858443

Country of ref document: EP

Effective date: 20210406

ENP Entry into the national phase

Ref document number: 112021004094

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210304

WWE Wipo information: entry into national phase

Ref document number: P-2024/0361

Country of ref document: RS